EP2249806A2 - Resveratrol formulations - Google Patents

Resveratrol formulations

Info

Publication number
EP2249806A2
EP2249806A2 EP09701250A EP09701250A EP2249806A2 EP 2249806 A2 EP2249806 A2 EP 2249806A2 EP 09701250 A EP09701250 A EP 09701250A EP 09701250 A EP09701250 A EP 09701250A EP 2249806 A2 EP2249806 A2 EP 2249806A2
Authority
EP
European Patent Office
Prior art keywords
peg
composition
formulation
resveratrol
alcohol
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP09701250A
Other languages
German (de)
French (fr)
Inventor
Walter Joseph Lunsmann
Peter Elliott
Philip Lambert
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sirtris Pharmaceuticals Inc
Original Assignee
Sirtris Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sirtris Pharmaceuticals Inc filed Critical Sirtris Pharmaceuticals Inc
Publication of EP2249806A2 publication Critical patent/EP2249806A2/en
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/05Phenols
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/06Free radical scavengers or antioxidants

Definitions

  • the Silent Information Regulator (SIR) family of genes represents a highly conserved group of genes present in the genomes of organisms ranging from archaebacteria to a variety of eukaryotes (Frye, 2000).
  • the encoded SIR proteins are involved in diverse processes from regulation of gene silencing to DNA repair.
  • the proteins encoded by members of the SIR2 gene family show high sequence conservation in a 250 amino acid core domain.
  • a well-characterized gene in this family is S. cerevisiae SIR2, which is involved in silencing HM loci that contain information specifying yeast mating type, telomere position effects and cell aging (Guarente, 1999; Kaeberlein et al., 1999; Shore, 2000).
  • the yeast Sir2 protein belongs to a family of histone deacetylases (reviewed in Guarente, 2000; Shore, 2000).
  • the Sir2 homolog, CobB in Salmonella typhimurium, functions as an NAD (nicotinamide adenine dinucleotide)-dependent ADP-ribosyl transferase (Tsang and Escalante-Semerena, 1998).
  • the Sir2 protein is a deacetylase which uses NAD as a cofactor (Imai et al., 2000; Moazed, 2001 ; Smith et al., 2000; Tanner et al., 2000; Tanny and Moazed, 2001 ). Unlike other deacetylases, many of which are involved in gene silencing, Sir2 is insensitive to histone deacetylase inhibitors like trichostatin A (TSA) (Imai et al., 2000; Landry et al., 2000a; Smith et al., 2000).
  • TSA histone deacetylase inhibitors
  • Deacetylation of acetyl-lysine by Sir2 is tightly coupled to NAD hydrolysis, producing nicotinamide and a novel acetyl-ADP ribose compound (1 -O-acetyl- ADP-ribose) (Tanner et al., 2000; Landry et al., 2000b; Tanny and Moazed, 2001 ).
  • the NAD-dependent deacetylase activity of Sir2 is essential for its functions which can connect its biological role with cellular metabolism in yeast (Guarente, 2000; Imai et al., 2000; Lin et al., 2000; Smith et al., 2000).
  • Sir2 homologs have NAD-dependent histone deacetylase activity (Imai et al., 2000; Smith et al., 2000). Most information about Sir2 mediated functions comes from the studies in yeast (Gartenberg, 2000; Gottschling, 2000).
  • small molecule activators of the SIR proteins such as resveratrol
  • small molecule activators of SIR proteins were shown to extend life span in yeast and cultured human cells as well as activate SIR protein activity in human cells ⁇ supra).
  • the small molecule SIR activators were shown to mimic calorie restriction and extend lifespan in Caenorhabditis elegans and Drosophila melanogaster ⁇ supra).
  • Activators of the SIR proteins may therefore be useful for mimicking the effects of calorie restriction in eukaryotic cells and treating aging-related diseases such as stroke, cardiovascular disease, arthritis, high blood pressure, or Alzheimer's disease ⁇ supra).
  • resveratrol promotes fat mobilization in C. elegans, prevent fat accumulation in C. elegans, stimulate fat mobilization in mammalian cells, and inhibit adipogenesis in mammalian cells (see e.g., U.S. Patent Publication No. 2005/0171027 and PCT Publication No. WO 2005/065667). Additionally, resveratrol was shown to at least partially restore insulin sensitivity in insulin resistant cells ⁇ supra).
  • resveratrol In order to attain the benefits seen in cells in a whole organism, it would be advantageous to have a formulation of resveratrol that could deliver a sufficient dose of the agent in a convenient form. In particular, it would be advantageous to have a highly concentrated liquid formulation of resveratrol, where the resveratrol stably remained in solution.
  • liquid formulations of resveratrol e.g., solutions, emulsions
  • liquid formulations of the invention include at least 1 1% or even 15% by weight of resveratrol.
  • the invention also includes methods of preparing these formulations.
  • the invention includes methods of treating a variety of conditions using the formulations described herein.
  • the invention includes the use of formulations of the invention for treating the conditions and diseases described herein, along with the use of the formulations in the manufacture of a medicament for treating the conditions and diseases described herein.
  • Figure 1 shows in vivo pharmacokinetic profiles of various resveratrol formulations administered to healthy human volunteers.
  • agent is used herein to denote a chemical compound, a mixture of chemical compounds, a biological macromolecule (such as a nucleic acid, an antibody, a protein or portion thereof, e.g., a peptide), or an extract made from biological materials such as bacteria, plants, fungi, or animal (particularly mammalian) cells or tissues.
  • a biological macromolecule such as a nucleic acid, an antibody, a protein or portion thereof, e.g., a peptide
  • an extract made from biological materials such as bacteria, plants, fungi, or animal (particularly mammalian) cells or tissues.
  • the activity of such agents may render it suitable as a "therapeutic agent” which is a biologically, physiologically, or pharmacologically active substance (or substances) that acts locally or systemically in a subject.
  • bioavailable when referring to a compound is art-recognized and refers to a form of a compound that allows for it, or a portion of the amount of compound administered, to be absorbed by, incorporated to, or otherwise physiologically available to a subject or patient to whom it is administered.
  • compact animals refers to cats and dogs.
  • dog(s) denotes any member of the species Canis familiaris, of which there are a large number of different breeds.
  • cat(s) refers to a feline animal including domestic cats and other members of the family Felidae, genus Felis.
  • Diabetes refers to high blood sugar or ketoacidosis, as well as chronic, general metabolic abnormalities arising from a prolonged high blood sugar status or a decrease in glucose tolerance. “Diabetes” encompasses both the type I and type II (Non Insulin Dependent Diabetes Mellitus or NIDDM) forms of the disease.
  • the risk factors for diabetes include the following factors: waistline of more than 40 inches for men or 35 inches for women, blood pressure of 130/85 mmHg or higher, triglycerides above 150 mg/dl, fasting blood glucose greater than 100 mg/dl or high- density lipoprotein of less than 40 mg/dl in men or 50 mg/dl in women.
  • hyperinsulinemia refers to a state in an individual in which the level of insulin in the blood is higher than normal.
  • insulin resistance refers to a state in which a normal amount of insulin produces a subnormal biologic response relative to the biological response in a subject that does not have insulin resistance.
  • insulin resistance disorder refers to any disease or condition that is caused by or contributed to by insulin resistance. Examples include: diabetes, obesity, metabolic syndrome, insulin-resistance syndromes, syndrome X, insulin resistance, high blood pressure, hypertension, high blood cholesterol, dyslipidemia, hyperlipidemia, dyslipidemia, atherosclerotic disease including stroke, coronary artery disease or myocardial infarction, hyperglycemia, hyperinsulinemia and/or hyperproinsulinemia, impaired glucose tolerance, delayed insulin release, diabetic complications, including coronary heart disease, angina pectoris, congestive heart failure, stroke, cognitive functions in dementia, retinopathy, peripheral neuropathy, nephropathy, glomerulonephritis, glomerulosclerosis, nephrotic syndrome, hypertensive nephrosclerosis some types of cancer (such as endometrial, breast, prostate, and colon), complications of pregnancy, poor female reproductive health (such as menstrual irregularities, infertility, irregular ovulation, poly
  • livestock animals refers to domesticated quadrupeds, which includes those being raised for meat and various byproducts, e.g., a bovine animal including cattle and other members of the genus Bos, a porcine animal including domestic swine and other members of the genus Sus, an ovine animal including sheep and other members of the genus Ovis, domestic goats and other members of the genus Capra; domesticated quadrupeds being raised for specialized tasks such as use as a beast of burden, e.g., an equine animal including domestic horses and other members of the family Equidae, genus Equus.
  • mammals include humans, primates, livestock animals (including bovines, porcines, etc.), companion animals (e.g., canines, felines, etc.) and rodents (e.g., mice and rats).
  • livestock animals including bovines, porcines, etc.
  • companion animals e.g., canines, felines, etc.
  • rodents e.g., mice and rats.
  • Obese individuals or individuals suffering from obesity are generally individuals having a body mass index (BMI) of at least 25 or greater. Obesity may or may not be associated with insulin resistance.
  • BMI body mass index
  • parenteral administration and “administered parenterally” are art-recognized and refer to modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intra- articulare, subcapsular, subarachnoid, intraspinal, and intrasternal injection and infusion.
  • a “patient”, “subject”, “individual” or “host” refers to either a human or a non-human animal.
  • pharmaceutically acceptable carrier is art-recognized and refers to a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting any subject composition or component thereof.
  • Each carrier must be “acceptable” in the sense of being compatible with the subject composition and its components and not injurious to the patient.
  • materials which may serve as pharmaceutically acceptable carriers include: (1 ) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (1 1 ) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum
  • phospholipids refers to lipids that contain a phosphate group, typically attached to the glycerol.
  • phospholipids include lecithin, hydroxylated lecithin, Lysophosphatidylcholine, Cardiolipin, Sphingomyelin, Phosphatidylcholine, Phosphatidyl ethanolamine, Phosphatidic acid, Phosphatidyl glycerol and Phosphatidyl serine.
  • polar lipid refers to a lipid having a full or partial positive or negative charge. Such lipids are typically surfactants.
  • prophylactic or therapeutic treatment refers to administration of a drug to a host. If it is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the host animal) then the treatment is prophylactic, i.e., it protects the host against developing the unwanted condition, whereas if administered after manifestation of the unwanted condition, the treatment is therapeutic (i.e., it is intended to diminish, ameliorate or maintain the existing unwanted condition or side effects therefrom).
  • Replicative lifespan of a cell refers to the number of daughter cells produced by an individual "mother cell.”
  • Increasing the lifespan of a cell or “extending the lifespan of a cell,” as applied to cells or organisms, refers to increasing the number of daughter cells produced by one cell; increasing the ability of cells or organisms to cope with stresses and combat damage, e.g., to DNA, proteins; and/or increasing the ability of cells or organisms to survive and exist in a living state for longer under a particular condition, e.g., stress (for example, heatshock, osmotic stress, high energy radiation, chemically-induced stress, DNA damage, inadequate salt level, inadequate nitrogen level, or inadequate nutrient level). Lifespan can he increased by at least about 20%, 30%, 40%, 50%, 60% or between 20% and 70%, 30% and 60%, 40% and 60% or more using methods described herein.
  • stress for example, heatshock, osmotic stress, high energy radiation, chemically-induced stress, DNA damage, inadequate salt level, inadequate nitrogen level, or inadequate nutrient level
  • sirtuin-activating compound refers to a compound that increases the level of a sirtuin protein and/or increases at least one activity of a sirtuin protein.
  • a sirtuin-activating compound may increase at least one biological activity of a sirtuin protein by at least about 10%, 25%, 50%, 75%, 100%, or more.
  • Exemplary biological activities of sirtuin proteins include deacetylation, e.g., of histones and p53; extending lifespan; increasing genomic stability; silencing transcription; and controlling the segregation of oxidized proteins between mother and daughter cells.
  • sirtuin protein refers to a member of the sirtuin deacetylase protein family, or preferably to the sir2 family, which include yeast Sir2 (GenBank Accession No. P53685), C. elegans Sir-2.1 (GenBank Accession No. NP_501912), and human SIRTl (GenBank Accession No. NMJ)12238 and NP_036370 (or AF083106)) and SIRT2 (GenBank Accession No. NMJ312237, NM_030593, NP_036369, NP_085096, and AF083107) proteins.
  • HST genes additional yeast Sir2-like genes termed "HST genes” (homologues of Sir two) HSTl , HST2, HST3 and HST4, and the five other human homologues hSIRT3, hSIRT4, hSIRT5, hSIRT6 and hSIRT7 (Brachmann et al. (1995) Genes Dev. 9:2888 and Frye et al. (1999) BBRC 260:273).
  • HST genes homologues of Sir two HSTl , HST2, HST3 and HST4
  • hSIRT3, hSIRT4, hSIRT5, hSIRT6 and hSIRT7 Bos et al. (1995) Genes Dev. 9:2888 and Frye et al. (1999) BBRC 260:273
  • SIRTl protein refers to a member of the sir2 family of sirtuin deacetylases.
  • a SIRTl protein includes yeast Sir2 (GenBank Accession No. P53685), C. elegans Sir-2.1 (GenBank Accession No. NP_501912), human SIRTl (GenBank Accession No. NM_O12238 or NP_036370 (or AF083106)), and human SIRT2 (GenBank Accession No. NM Ol 2237, NM_030593, NP 036369,
  • a SIRTl protein includes a polypeptide comprising a sequence consisting of, or consisting essentially of, the amino acid sequence set forth in GenBank Accession Nos. NP 036370, NP_501912, NP_085096, NP 036369, or P53685.
  • SIRTl proteins include polypeptides comprising all or a portion of the amino acid sequence set forth in GenBank Accession Nos. NP 036370, NP 501912, NPJD85096, NP 036369, or P53685; the amino acid sequence set forth in GenBank Accession Nos.
  • Polypeptides of the invention also include homologs (e.g., orthologs and paralogs), variants, or fragments, of GenBank Accession Nos. NP 036370, NP_501912, NP_085096, NP_036369, or P53685.
  • a surfactant is a compound having both polar (hydrophilic) and nonpolar (hydrophobic) regions.
  • the polar region of a surfactant may be ionic (positive or negative) or nonionic.
  • Surfactants are useful in preparing emulsions and in solubilizing substances.
  • systemic administration "administered systemically,”
  • peripheral administration and “administered peripherally” are art-recognized and refer to the administration of a subject composition, therapeutic or other material other than directly into the central nervous system, such that it enters the patient's system and, thus, is subject to metabolism and other like processes.
  • therapeutic agent is art-recognized and refers to any chemical moiety that is a biologically, physiologically, or pharmacologically active substance that acts locally or systemically in a subject.
  • the term also means any substance intended for use in the diagnosis, cure, mitigation, treatment or prevention of disease or in the enhancement of desirable physical or mental development and/or conditions in an animal or human.
  • therapeutic effect is art-recognized and refers to a local or systemic effect in animals, particularly mammals, and more particularly humans caused by a pharmacologically active substance.
  • therapeutically- effective amount means that amount of such a substance that produces some desired local or systemic effect at a reasonable benefit/risk ratio applicable to any treatment.
  • the therapeutically effective amount of such substance will vary depending upon the subject and disease condition being treated, the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
  • certain compositions described herein may be administered in a sufficient amount to produce a desired effect at a reasonable benefit/risk ratio applicable to such treatment.
  • Treating refers to curing as well as ameliorating at least one symptom of the condition or disease.
  • vision impairment refers to diminished vision, which is often only partially reversible or irreversible upon treatment (e.g., surgery). Particularly severe vision impairment is termed “blindness” or “vision loss”, which refers to a complete loss of vision, vision worse than 20/200 that cannot be improved with corrective lenses, or a visual field of less than 20 degrees diameter (10 degrees radius).
  • the invention provides novel pharmaceutical compositions (also referred to as "formulations") for treating and/or preventing a wide variety of diseases and disorders including, for example, diseases or disorders related to aging or stress, diabetes, obesity, neurodegenerative diseases, ocular diseases and disorders, cardiovascular diseases, blood clotting disorders, inflammation, cancer, and/or flushing, etc.
  • Pharmaceutical compositions of the invention may also be used for treating a disease or disorder in a subject that would benefit from increased mitochondrial activity, for enhancing muscle performance, for increasing muscle ATP levels, or for treating or preventing muscle tissue damage associated with hypoxia or ischemia.
  • Resveratrol is 3,5,4'-trihydroxystilbene and has both cis and trans isomers.
  • the structure of each isomer is shown below:
  • the invention also includes salts and esters of resveratrol, where indicated.
  • Base addition salts include those derived from inorganic bases, such as ammonium or alkali or alkaline earth metal hydroxides, carbonates, bicarbonates, and the like.
  • bases useful in preparing the salts of this invention thus include sodium hydroxide, potassium hydroxide, ammonium hydroxide, potassium carbonate, and the like.
  • the compounds and salts and esters thereof can be present in amorphous or crystalline (including co-crystalline and polymorph) forms, unless otherwise indicated.
  • the liquid formulations of the invention include at least 1 1% by weight of resveratrol or a salt or ester thereof.
  • the formulation includes at least 12%, at least 13%, at least 14%, at least 15%, at least 16%, at least 17%, at least 1 8%, at least 19%, at least 20%, at least 21 %, at least 22%, at least 23%, at least 24% or at least 25% of resveratrol (or its salt or ester).
  • the formulation includes resveratrol and one or more of monoglycerides, diglycerides, triglycerides, fatty acid monoesters of polyethylene glycol (PEG) and fatty acid diesters of PEG.
  • the monoglycerides, diglycerides, triglycerides, fatty acid monoesters of PEG and fatty acid diesters of PEG include C8 and C l O fatty acids, such as caprylocapryol macrogol glycerides.
  • the formulation is a solution and the solution consists essentially of resveratrol and caprylocapryol macrogol glycerides (commercially available as LabrasolTM).
  • Such formulations may also include a polar lipid, such as a phospholipid (e.g., lecithin).
  • a polar lipid such as a phospholipid (e.g., lecithin).
  • the amount of polar lipid is typically from 0.25-5% (e.g., by weight), such as 1-3% or 1.5-2.5%, with the remainder resveratrol and monoglycerides, diglycerides, triglycerides, fatty acid monoesters of polyethylene glycol (PEG) and fatty acid diesters of PEG.
  • PEG polyethylene glycol
  • such formulations may also include one or more of saturated polyglycolized glycerides (e.g., Gelucire 44/14) and ethanol.
  • saturated polyglycolized glycerides e.g., Gelucire 44/114
  • ethanol e.g., ethanol
  • the amount of saturated polyglycolized glycerides and/or ethanol is no greater than 20%, such as 1 -15%, 1 -10% or 1 -5% by weight.
  • the formulation comprises a triglyceride, a propylene glycol fatty acid ester, a polyethylene glycol sorbitan fatty acid ester, a saturated polyglycolized glyceride and an alcohol.
  • the triglyceride is a long chain triglyceride
  • the propylene glycol fatty acid ester is propylene glycol caprylate/caprate
  • the polyethylene glycol sorbitan fatty acid ester is polyoxyethylated sorbitan monooleate
  • the saturated polyglycolized glyceride is lauryol macrogol glycerides
  • the alcohol is ethanol.
  • the formulation contains 1 -15% (e.g., 5-10%) by weight of the triglyceride, 10-25% (e.g., 15-20%) by weight of the propylene glycol fatty acid ester, 25-45% (e.g. 30- 40%) by weight of the polyethylene glycol sorbitan fatty acid ester, 10-25% (e.g. 15- 20%) by weight of the saturated polyglycolized glyceride and 5-20% (e.g., 10-15%) by weight of the alcohol.
  • the formulation comprises resveratrol, a monoacyl phospholipid, a PEG and a dihydric alcohol.
  • the PEG is PEG 300 and the dihydric alcohol is propylene glycol.
  • the formulation contains 50-70% (e.g., 55-65%) by weight of the monoacyl phospholipid, 10-25% (e.g., 15-20%) by weight of the PEG and 10-25% (e.g., 15-20%) by weight of the dihydric alcohol
  • the formulation comprises resveratrol, a polyethoxylated fatty acid, an alcohol-oil transesterification product and an alcohol.
  • the polyethoxylated fatty acid is polyethoxylated glycol hydroxystearate
  • the alcohol-oil transesterification product is D-alpha-tocopheryl PEG 1000 succinate
  • the alcohol is ethanol.
  • the formulation contains 30-50% (e.g., 35-45%) by weight of the polyethoxylated fatty acid, 1-15% (e.g., 3- 8%) by weight of the alcohol-oil transesterification product and 40-60% (e.g., 45- 55%) by weight, of the alcohol.
  • the formulation comprises resveratrol, a triglyceride, an alcohol-oil transesterification product, a monoglyceride and an alcohol.
  • the triglyceride is a medium chain triglyceride
  • the alcohol-oil transesterification product is a polyethxoylated hydrogenated castor oil
  • the monoglyceride is an oleate
  • the alcohol is ethanol.
  • the formulation contains 15-30% (e.g., 20-25%) by weight of the triglyceride, 35- 55% (e.g., 40-50%) by weight of the alcohol-oil transesterification product, 1-15% (e.g., 3-8%) by weight of the monoglyceride and 15-30% (e.g., 20-25%) by weight of the alcohol.
  • the formulation comprises a polymer and a surfactant. Suitable polymers include polymers that have a full or partial positive charge, such as amine-containing polymers. An example of such a polymer is polyvinylpyrrolidone and derivatives thereof. Other examples include polyallylamine and polyvinylamine and derivatives thereof.
  • Saturated polyglycolized glycerides are triglycerides that have been partially hydrolyzed with PEG, thereby forming a mixture of monoglycerides, diglycerides, triglycerides, fatty acid monoesters of PEG and fatty acid diesters of PEG. Examples of such compositions include Gelucire 44/14, Gelucire 50/13 and Gelucire 53/10. Monoglycerides are fatty acid monoesters of glycerol.
  • monoglycerides examples include glyceryl monooleate, glyceryl 15 ricinoleate, glyceryl laurate, glyceryl monooleate, glyceryl caprylate/caprate, caprylic acid monoglycerides, and monoacetylated monoglycerides.
  • Triglycerides are fatty acid triesters of glycerol.
  • examples of triglycerides include Aceituno oil, Almond oil, Super Refined Almond Oil, Araehis oil, Babassu oil, Blackcurrant seed oil, Borage oil, Buffalo ground oil, Candlenut oil, Canola oil, Lipex 108, Castor oil, Chinese vegetable tallow oil, Cocoa butter, coconut oil, Pureco 76, Coffee seed oil, Corn oil, Super Refined Corn Oil, Cottonseed oil, Super Refined Cottonseed Oil, Crambe oil, Cuphea species oil, Evening primrose oil, Grapeseed oil, Groundnut oil, Hemp seed oil, Illipe butter, Kapok seed oil, Linseed oil, Menhaden oil, Super Refined Menhaden Oil, Mowrah butter, Mustard seed oil, Oiticica oil, Olive oil, Super Refined Olive Oil, Palm oil, Palm kernel oil, Peanut oil, Super Refined Peanut Oil, Poppy seed oil, Rapeseed
  • Fractionated triglycerides, modified triglycerides, synthetic triglycerides, and mixtures of triglycerides are considered triglycerides for this invention.
  • a medium-chain triglyceride typically has 8 to 10 carbon atoms, whereas a long-chain triglyceride has 12 or more carbon atoms (typically 12-14, such as more than 16 carbon atoms is referred to as a very long chain).
  • Diglycerides have only two fatty acid moieties attached to glycerol.
  • Monoacyl phospholipids include a hydrocarbon chain portion and a phospholipid portion.
  • the hydrocarbon chain can either be unsaturated or saturated and can have between 10 to 24, preferably 14 to 18 carbon atoms.
  • the lipids may be derived from natural plant, or animal or microbiological sources (e.g., phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylinositol (PI), phosphatidic acid (PI) and/or phosphatidylserine (PS)), synthesized or partially synthesized, including polyethyleneglycol (PEG) derived-monoacyl phospholipids (e.g., pegylated monoacyl phosphatidyl ethanolamine).
  • Exemplary monoacyl phospholipids include enzyme modified (Phospholipase A2) soy PC, Egg PC, 1 - palmitoyl PC, 1 oleoyl PC, and 1 -stearoyl PC.
  • propylene glycol fatty acid esters examples include Propylene glycol monocaprylate, Propylene glycol monolaurate, Propylene glycol oleate, Propylene glycol myristate, Propylene glycol monostearate, Propylene glycol hydroxy stearate, Propylene glycol ricinoleate, Propylene glycol isostearate, Propylene glycol monooleate, Propylene glycol dicaprylate/dicaprate, Propylene glycol dioctanoate, Propylene glycol caprylate/caprate, Propylene glycol dilaurate, Propylene glycol distearate, Propylene glycol dicaprylate, and Propylene glycol dicaprate.
  • polyethylene glycol sorbitan fatty acid esters examples include PEG-10 sorbitan laurate, PEG-20 sorbitan monolaurate, PEG-4 sorbitan monolaurate, PEG- 80 sorbitan monolaurate, PEG-6 sorbitan monolaurate, PEG-20 sorbitan monopalmitate, PEG-20 sorbitan monostearate, PEG-4 sorbitan monostearate, PEG- 8 sorbitan monostearate, PEG-6 sorbitan monostearate, PEG-20 sorbitan tristearate, PEG-6 sorbitan tetrastearate, PEG-60 sorbitan tetrastearate, PEG-5 sorbitan monooleate, PEG-6 sorbitan monooleate, PEG-20 sorbitan monooleate, PEG-40 sorbitan oleate, PEG-20 sorbitan trioleate, PEG-6 sorbitan tetraoleate, PEG-30
  • Examples of monohydric alcohols include methanol, ethanol, isopropanol and n-propanol. Typically, the alcohol is ethanol.
  • Dihydric alcohols include two hydroxyl groups, such as ethylene glycol and propylene glycol.
  • Polyethylene glycols can have any suitable length, such as PEG- 100, PEG- 200, PEG-300, PEG-400, PEG-500, PEG- 1000 and so forth.
  • polyethoxylated fatty acids examples include PEG 4- 100 monolaurate, PEG 4- 100 monooleate, PEG 4-100 monostearate, PEG 400 distearate, PEG
  • a large number of surfactants of different degrees of hydrophobicity or hydrophilicity can be prepared by reaction of alcohols or polyalcohols with a variety of natural and/or hydrogenated oils.
  • the oils used are castor oil or hydrogenated castor oil, or an edible vegetable oil such as com oil, olive oil, peanut oil, palm kernel oil, apricot kernel oil, or almond oil.
  • Suitable alcohols include alcohols include glycerol, propylene glycol, ethylene glycol, polyethylene glycol, sorbitol, and pentaerythritol.
  • alcohol-oil transesterification products include PEG-3 castor oil, PEG-5, 9, and 16 castor oil, PEG-20 castor oil, PEG-23 castor oil, PEG-30 castor oil, PEG-35 castor oil, PEG-38 castor oil, PEG-40 castor oil, PEG-50 castor oil, PEG-56 castor oil, PEG-60 castor oil, PEG-100 castor oil, PEG-200 castor oil, PEG-5 hydrogenated castor oil, PEG-7 hydrogenated castor oil, PEG- 10 hydrogenated castor oil, PEG-20 hydrogenated castor oil, PEG-25 hydrogenated castor oil, PEG-30 hydrogenated castor oil, PEG-40 hydrogenated castor oil, PEG-45 hydrogenated castor oil, PEG-50 hydrogenated castor oil, PEG- 60 hydrogenated castor oil, PEG-80 hydrogenated castor oil, PEG-100 hydrogenated castor oil, PEG-6 corn oil, PEG-6 almond oil, PEG-6 apricot kernel oil, PEG-6 olive oil, PEG
  • the invention provides methods for modulating the level and/or activity of a sirtuin protein and methods of use thereof.
  • the invention provides methods for using formulations of the invention wherein the formulations activate a sirtuin protein, e.g., increase the level and/or activity of a sirtuin protein.
  • Formulations that increase the level and/or activity of a sirtuin protein may be useful for a variety of therapeutic applications including, for example, increasing the lifespan of a cell, and treating and/or preventing a wide variety of diseases and disorders including, for example, diseases or disorders related to aging or stress, diabetes, obesity, neurodegenerative diseases, cardiovascular disease, blood clotting disorders, inflammation, cancer, and/or flushing, etc.
  • the methods comprise administering to a subject in need thereof a pharmaceutically effective amount of a formulation.
  • the formulations described herein may be taken alone or in combination with other compounds.
  • a mixture of resveratrol and one or more other sirtuin-modulating compounds may be administered to a subject in need thereof.
  • a formulation may be administered with one or more of the following compounds: butein, fisetin, piceatannol, or quercetin.
  • a formulation may be administered in combination with nicotinic acid.
  • formulations may be administered with one or more therapeutic agents for the treatment or prevention of various diseases, including, for example, cancer, diabetes, neurodegenerative diseases, cardiovascular disease, blood clotting, inflammation, flushing, obesity, ageing, stress, etc.
  • combination therapies comprising a formulation of the invention may refer to ( 1 ) pharmaceutical compositions that comprise one or more formulations in combination with one or more therapeutic agents (e.g., one or more therapeutic agents described herein); and (2) co-administration of one or more formulations with one or more therapeutic agents wherein the formulation of the invention and therapeutic agent have not been formulated in the same compositions (but may be present within the same kit or package, such as a blister pack or other multi-chamber package; connected, separately sealed containers (e.g., foil pouches) that can be separated by the user; or a kit where the formulation of the invention and other therapeutic agent(s) are in separate vessels).
  • the formulation of the invention may be administered at the same, intermittent, staggered, prior to, subsequent to,
  • methods for reducing, preventing or treating diseases or disorders using a formulation of the invention may also comprise increasing the protein level of a sirtuin, such as human SIRTl , SIRT2 and/or SIRT3, or homologs thereof.
  • a sirtuin such as human SIRTl , SIRT2 and/or SIRT3, or homologs thereof.
  • Increasing protein levels can be achieved by introducing into a cell one or more copies of a nucleic acid that encodes a sirtuin.
  • the level of a sirtuin can be increased in a mammalian cell by introducing into the mammalian cell a nucleic acid encoding the sirtuin, e.g., increasing the level of SIRTl by introducing a nucleic acid encoding the amino acid sequence set forth in GenBank Accession No.
  • NP_036370 and/or increasing the level of SIRT3 by introducing a nucleic acid encoding the amino acid sequence set forth in GenBank Accession No. AAH01042.
  • the nucleic acid may be under the control of a promoter that regulates the expression of the SIRTl and/or SIRT3 nucleic acid.
  • the nucleic acid may be introduced into the cell at a location in the genome that is downstream of a promoter. Methods for increasing the level of a protein using these methods are well known in the art.
  • a nucleic acid that is introduced into a cell to increase the protein level of a sirtuin may encode a protein that is at least about 80%, 85%, 90%, 95%, 98%, or 99% identical to the sequence of a sirtuin, e.g., SIRTl (GenBank Accession No. NP 036370) and/or SIRT3 (GenBank Accession No. AAHO 1042) protein.
  • the nucleic acid encoding the protein may be at least about 80%, 85%, 90%, 95%, 98%, or 99% identical to a nucleic acid encoding a SIRTl (e.g. GenBank Accession No.
  • the nucleic acid may also be a nucleic acid that hybridizes, preferably under stringent hybridization conditions, to a nucleic acid encoding a wild-type sirtuin, e.g., SIRTl (GenBank Accession No. NM_012238) and/or SIRT3 (e.g., GenBank Accession No. BCOOl 042) protein.
  • Stringent hybridization conditions may include hybridization and a wash in 0.2 x SSC at 65 0 C.
  • a protein that is different from a wild-type sirtuin protein such as a protein that is a fragment of a wild-type sirtuin
  • the protein is preferably biologically active, e.g., is capable of deacetylation. It is only necessary to express in a cell a portion of the sirtuin that is biologically active.
  • a protein that differs from wild-type SIRTl having GenBank Accession No. NP 036370 preferably contains the core structure thereof.
  • the core structure sometimes refers to amino acids 62-293 of GenBank Accession No. NP 036370, which are encoded by nucleotides 237 to 932 of GenBank Accession No.
  • the core domain of SIRTl may also refer to about amino acids 261 to 447 of GenBank Accession No. NP 036370, which are encoded by nucleotides 834 to 1394 of GenBank Accession No. NM 012238; to about amino acids 242 to 493 of GenBank Accession No. NP 036370, which are encoded by nucleotides 777 to 1532 of GenBank Accession No. NM 012238; or to about amino acids 254 to 495 of
  • methods for reducing, preventing or treating diseases or disorders using a formulation of the invention may also comprise decreasing the protein level of a sirtuin, such as human SIRTl , SIRT2 and/or SIRT3, or homologs thereof. Decreasing a sirtuin protein level can be achieved according to methods known in the art.
  • an siRNA, an antisense nucleic acid, or a ribozyme targeted to the sirtuin can be expressed in the cell.
  • a dominant negative sirtuin mutant e.g., a mutant that is not capable of deacetylating, may also be used.
  • mutant H363Y of SIRTl described, e.g., in Luo et al. (2001 ) Cell 107: 137 can be used.
  • agents that inhibit transcription can be used.
  • Methods for modulating sirtuin protein levels also include methods for modulating the transcription of genes encoding sirtuins, methods for stabilizing/destabilizing the corresponding mRNAs, and other methods known in the art. Aging/Stress
  • the invention provides a method extending the lifespan of a cell, extending the proliferative capacity of a cell, slowing ageing of a cell, promoting the survival of a cell, delaying cellular senescence in a cell, mimicking the effects of calorie restriction, increasing the resistance of a cell to stress, or preventing apoptosis of a cell, by contacting the cell with a formulation of the invention that increases the level and/or activity of a sirtuin protein.
  • the methods described herein may be used to increase the amount of time that cells, particularly primary cells (i.e., cells obtained from an organism, e.g., a human), may be kept alive in a cell culture.
  • Embryonic stem (ES) cells and pluripotent cells, and cells differentiated therefrom may also be treated with a - formulation of the invention to keep the cells, or progeny thereof, in culture for longer periods of time.
  • Such cells can also be used for transplantation into a subject, e.g., after ex vivo modification.
  • cells that are intended to be preserved for long periods of time may be treated with a formulation of the invention.
  • the cells may be in suspension (e.g.. blood cells, serum, biological growth media, etc.) or in tissues or organs.
  • blood collected from an individual for purposes of transfusion may be treated with a formulation of the invention to preserve the blood cells for longer periods of time.
  • blood to be used for forensic purposes may also be preserved using a formulation of the invention.
  • Other cells that may be treated to extend their lifespan or protect against apoptosis include cells for consumption, e.g., cells from non-human mammals (such as meat) or plant cells (such as vegetables).
  • Formulations of the invention may also be applied during developmental and growth phases in mammals, plants, insects or microorganisms, in order to, e.g., alter, retard or accelerate the developmental and/or growth process.
  • formulations of the invention may be used to treat cells useful for transplantation or cell therapy, including, for example, solid tissue grafts, organ transplants, cell suspensions, stem cells, bone marrow cells, etc.
  • the cells or tissue may be an autograft, an allograft, a syngraft or a xenograft.
  • the cells or tissue may be treated with the formulation of the invention prior to administration/implantation, concurrently with administration/implantation, and/or post administration/implantation into a subject.
  • the cells or tissue may be treated prior to removal of the cells from the donor individual, ex vivo after removal of the cells or tissue from the donor individual, or post implantation into the recipient.
  • the donor or recipient individual may be treated systemically with a formulation of the invention or may have a subset of cells/tissue treated locally with a formulation of the invention.
  • the cells or tissue may additionally be treated with another therapeutic agent useful for prolonging graft survival, such as, for example, an immunosuppressive agent, a cytokine, an angiogenic factor, etc.
  • cells may be treated with a formulation of the invention in vivo, e.g., to increase their lifespan or prevent apoptosis.
  • skin can be protected from aging (e.g., developing wrinkles, loss of elasticity, etc.) by treating skin or epithelial cells with a formulation of the invention.
  • skin is contacted with a formulation of the invention.
  • Exemplary skin afflictions or skin conditions that may be treated in accordance with the methods described herein include disorders or diseases associated with or caused by inflammation, sun damage or natural aging.
  • compositions find utility in the prevention or treatment of contact dermatitis (including irritant contact dermatitis and allergic contact dermatitis), atopic dermatitis (also known as allergic eczema), actinic keratosis, keratinization disorders (including eczema), epidermolysis bullosa diseases (including penfigus), exfoliative dermatitis, seborrheic dermatitis, erythemas (including erythema multiforme and erythema nodosum), damage caused by the sun or other light sources, discoid lupus erythematosus, dermatomyositis, psoriasis, skin cancer and the effects of natural aging.
  • contact dermatitis including irritant contact dermatitis and allergic contact dermatitis
  • atopic dermatitis also known as allergic eczema
  • actinic keratosis also known as allergic eczema
  • a formulation of the invention may be used for the treatment of wounds and/or burns to promote healing, including, for example, first-, second- or third-degree burns and/or thermal, chemical or electrical burns.
  • the formulations may be administered topically, to the skin or mucosal tissue, as an ointment, lotion, cream, microemulsion, gel, solution or the like, as further described herein, within the context of a dosing regimen effective to bring about the desired result.
  • Topical formulations may also be used as preventive, e.g., chemopreventive, compositions.
  • preventive e.g., chemopreventive
  • susceptible skin is treated prior to any visible condition in a particular individual.
  • Formulations of the invention may be delivered locally or systemically to a subject.
  • a formulation of the invention is delivered locally to a tissue or organ of a subject by injection, topical formulation, etc.
  • a formulation of the invention may be used for treating or preventing a disease or condition induced or exacerbated by cellular senescence in a subject; methods for decreasing the rate of senescence of a subject, e.g., after onset of senescence; methods for extending the lifespan of a subject; methods for treating or preventing a disease or condition relating to lifespan; methods for treating or preventing a disease or condition relating to the proliferative capacity of cells; and methods for treating or preventing a disease or condition resulting from cell damage or death.
  • the method does not act by decreasing the rate of occurrence of diseases that shorten the lifespan of a subject.
  • a method does not act by reducing the lethality caused by a disease, such as cancer.
  • a formulation of the invention may be administered to a subject in order to generally increase the lifespan of its cells and to protect its cells against stress and/or against apoptosis. It is believed that treating a subject with a compound described herein is similar to subjecting the subject to ho ⁇ nesis, i.e., mild stress that is beneficial to organisms and may extend their lifespan. Formulations of the invention may be administered to a subject to prevent aging and aging-related consequences or diseases, such as stroke, heart disease, heart failure, arthritis, high blood pressure, and Alzheimer's disease. Other conditions that can be treated include ocular disorders, e.g., associated with the aging of the eye, such as cataracts, glaucoma, and macular degeneration.
  • Formulations of the invention can also be administered to subjects for treatment of diseases, e.g., chronic diseases, associated with cell death, in order to protect the cells from cell death.
  • diseases include those associated with neural cell death, neuronal dysfunction, or muscular cell death or dysfunction, such as Parkinson's disease, Alzheimer's disease, multiple sclerosis, amniotropic lateral sclerosis, and muscular dystrophy; AIDS; fulminant hepatitis; diseases linked to degeneration of the brain, such as Creutzfeld-Jakob disease, retinitis pigmentosa and cerebellar degeneration; myelodysplasis such as aplastic anemia; ischemic diseases such as myocardial infarction and stroke; hepatic diseases such as alcoholic hepatitis, hepatitis B and hepatitis C; joint diseases such as osteoarthritis; atherosclerosis; alopecia; damage to the skin due to UV light; lichen planus; atrophy of the skin; cataract; and graft rejection
  • Formulation of the inventions can also be administered to a subject suffering from an acute disease, e.g., damage to an organ or tissue, e.g., a subject suffering from stroke or myocardial infarction or a subject suffering from a spinal cord injury.
  • Formulations of the invention may also be used to repair an alcoholic's liver.
  • Car dio vascular Disease In another embodiment, the invention provides a method for treating and/or preventing a cardiovascular disease by administering to a subject in need thereof a formulation of the invention.
  • Cardiovascular diseases that can be treated or prevented using the formulations of the invention include cardiomyopathy or myocarditis; such as idiopathic cardiomyopathy, metabolic cardiomyopathy, alcoholic cardiomyopathy, drug-induced cardiomyopathy, ischemic cardiomyopathy, and hypertensive cardiomyopathy.
  • cardiomyopathy or myocarditis such as idiopathic cardiomyopathy, metabolic cardiomyopathy, alcoholic cardiomyopathy, drug-induced cardiomyopathy, ischemic cardiomyopathy, and hypertensive cardiomyopathy.
  • atheromatous disorders of the major blood vessels such as the aorta, the coronary arteries, the carotid arteries, the cerebrovascular arteries, the renal arteries, the iliac arteries, the femoral arteries, and the popliteal arteries.
  • vascular diseases that can be treated or prevented include those related to platelet aggregation, the retinal arterioles, the glomerular arterioles, the vasa nervorum, cardiac arterioles, and associated capillary beds of the eye, the kidney, the heart, and the central and peripheral nervous systems.
  • the formulations of the invention may also be used for increasing HDL levels in plasma of an individual.
  • disorders that may be treated with a formulation of the invention include restenosis, e.g., following coronary intervention, and disorders relating to an abnormal level of high density and low density cholesterol.
  • a formulation of the invention may be administered as part of a combination therapeutic with another cardiovascular agent including, for example, an anti-arrhythmic agent, an antihypertensive agent, a calcium channel blocker, a cardioplegic solution, a cardiotonic agent, a fibrinolytic agent, a sclerosing solution, a vasoconstrictor agent, a vasodilator agent, a nitric oxide donor, a potassium channel blocker, a sodium channel blocker, statins, or a natriuretic agent.
  • another cardiovascular agent including, for example, an anti-arrhythmic agent, an antihypertensive agent, a calcium channel blocker, a cardioplegic solution, a cardiotonic agent, a fibrinolytic agent, a sclerosing solution, a vasoconstrictor agent, a vasodilator agent, a nitric oxide donor, a potassium channel blocker, a sodium channel blocker, statins, or
  • a formulation of the invention may be administered as part of a combination therapeutic with an anti-arrhythmia agent.
  • Anti-arrhythmia agents are often organized into four main groups according to their mechanism of _. action: type I, sodium channel blockade; type II, beta-adrenergic blockade; type 111, repolarization prolongation; and type IV, calcium channel blockade.
  • Type I antiarrhythmic agents include lidocaine, moricizine, mexiletine, tocainide, procainamide, encainide, flecanide, tocainide, phenytoin, propafenone, quinidine, disopyramide, and flecainide.
  • Type II anti-arrhythmic agents include propranolol and esmolol.
  • Type III includes agents that act by prolonging the duration of the action potential, such as amiodarone, artilide, bretylium, clofilium, isobutilide, sotalol, azimilide, dofetilide, dronedarone, ersentilide, ibutilide, tedisamil, and VEtilide.
  • Type IV anti-arrhythmic agents include verapamil, diltaizem, digitalis, adenosine, nickel chloride, and magnesium ions.
  • a formulation of the invention may be administered as part of a combination therapeutic with another cardiovascular agent.
  • cardiovascular agents examples include vasodilators, for example, hydralazine; angiotensin converting enzyme inhibitors, for example, captopril; anti-anginal agents, for example, isosorbide nitrate, glyceryl trinitrate and pentaerythritol tetranitrate; antiarrhythmic agents, for example, quinidine, procainaltide and lignocaine; cardioglycosides, for example, digoxin and digitoxin; calcium antagonists, for example, verapamil and nifedipine; diuretics, such as thiazides and related compounds, for example, bendrofluazide, chlorothiazide, chlorothalidone, hydrochlorothiazide and other diuretics, for example, fursemide and triamterene, and sedatives, for example, nitrazepam, flurazepam and diazepam.
  • vasodilators for example,
  • exemplary cardiovascular agents include, for example, a cyclooxygenase inhibitor such as aspirin or indomethacin, a platelet aggregation inhibitor such as clopidogrel, ticlopidene or aspirin, fibrinogen antagonists or a diuretic such as chlorothiazide, hydrochlorothiazide, flumethiazide, hydroflumethiazide, bendro flumethiazide, methyl chlorthiazide, trichloromethiazide, polythiazide or benzthiazide as well as ethacrynic acid tricrynafen, chlorthalidone, furosemide, musolimine, bumetanide, triamterene, amiloride and spironolactone and salts of such compounds, angiotensin converting enzyme inhibitors such as captopril, zofenopril, fosinopril, enalapril, ceranopril,
  • cardiovascular agents include, for example, vasodilators, e.g., bencyclane, cinnarizine, citicoline, cyclandelate, cyclonicate, ebumamonine, phenoxezyl, flunarizine, ibudilast, ifenprodil, lomerizine, naphlole, nikamate, nosergoline, nimodipine, papaverine, pentifylline, nofedoline, vincamin, vinpocetine, vichizyl, pentoxifylline, prostacyclin derivatives (such as prostaglandin El and prostaglandin 12), an endothelin receptor blocking drug (such as bosentan), diltiazem, nicorandil, and nitroglycerin.
  • vasodilators e.g., bencyclane, cinnarizine, citicoline, cyclandelate, cyclonicate, e
  • Examples of the cerebral protecting drug include radical scavengers (such as edaravone, vitamin E, and vitamin C), glutamate antagonists, AMPA antagonists, kainate antagonists, NMDA antagonists, GABA agonists, growth factors, opioid antagonists, phosphatidylcholine precursors, serotonin agonists, Na + /Ca 2+ channel inhibitory drugs, and K + channel opening drugs.
  • Examples of the brain metabolic stimulants include amantadine, tiapride, and gamma-aminobutyric acid.
  • anticoagulant examples include heparins (such as heparin sodium, heparin potassium, dalteparin sodium, dalteparin calcium, heparin calcium, parnaparin sodium, reviparin sodium, and danaparoid sodium), warfarin, enoxaparin, argatroban, batroxobin, and sodium citrate.
  • heparins such as heparin sodium, heparin potassium, dalteparin sodium, dalteparin calcium, heparin calcium, parnaparin sodium, reviparin sodium, and danaparoid sodium
  • warfarin warfarin
  • enoxaparin argatroban
  • batroxobin and sodium citrate.
  • antiplatelet drug examples include ticlopidine hydrochloride, dipyridamole, cilostazol, ethyl icosapentate, sarpogrelate hydrochloride, dilazep hydrochloride, trapidil, a nonsteroidal antiinflammatory agent (such as aspirin), beraprostsodium, iloprost, and indobufene.
  • thrombolytic drug include urokinase, tissue-type plasminogen activators (such as alteplase, tisokinase, nateplase, pamiteplase, monteplase, and rateplase), and nasaruplase.
  • antihypertensive drug examples include angiotensin converting enzyme inhibitors (such as captopril, alacepril, lisinopril, imidapril, quinapril, temocapril, delapril, benazepril, cilazapril, trandolapril, enalapril, ceronapril, fosinopril, imadapril, mobertpril, perindopril, ramipril, spirapril, and randolapril), angiotensin 11 antagonists (such as losartan, candesartan, valsartan, eprosartan, and irbesartan), calcium channel blocking drugs (such as aranidipine, efonidipine, nicardipine, bamidipine, benidipine, manidipine, cilnidipine, nisoldipine, nitrendipin
  • antianginal drug examples include nitrate drugs (such as amyl nitrite, nitroglycerin, and isosorbide), ⁇ -adrenaline receptor blocking drugs (such as propranolol, pindolol, indenolol, carteolol, bunitrolol, atenolol, acebutolol, metoprolol, timolol, nipradilol, penbutolol, nadolol, tilisolol, carvedilol, bisoprolol, betaxolol, celiprolol, bopindolol, bevantolol, labetalol, alprenolol, amosulalol, arotinolol, befunolol, bucumolol, bufetolol, buferalol, buprandolol, butylidine,
  • diuretic examples include thiazide diuretics (such as hydrochlorothiazide, methyclothiazide, trichlormethiazide, benzylhydrochlorothiazide, and penflutizide), loop diuretics (such as furosemide, etacrynic acid, bumetanide, piretanide, azosemide, and torasemide), K + sparing diuretics (spironolactone, triamterene, andpotassiumcanrenoate), osmotic diuretics (such as isosorbide, D-mannitol, and glycerin), nonthiazide diuretics (such as meticrane, tripamide, chlorthalidone, and mefruside), and acetazolamide.
  • thiazide diuretics such as hydrochlorothiazide, methyclothiazide, trichlormethiazide, benzylhydr
  • cardiotonic examples include digitalis formulations (such as digitoxin, digoxin, methyldigoxin, deslanoside, vesnarinone, lanatoside C, and proscillaridin), xanthine formulations (such as aminophylline, choline theophylline, diprophylline, and proxyphylline), catecholamine formulations (such as dopamine, dobutamine, and docarpamine), PDE III inhibitors (such as amrinone, olprinone, and milrinone), denopamine, ubidecarenone, pimobendan, levosimendan, aminoethylsulfonic acid, vesnarinone, carperitide, and colforsin daropate.
  • digitalis formulations such as digitoxin, digoxin, methyldigoxin, deslanoside, vesnarinone, lanatoside C, and proscillaridin
  • xanthine formulations such
  • antiarrhythmic drug examples include ajmaline, pirmenol, procainamide, cibenzoline, disopyramide, quinidine, aprindine, mexiletine, lidocaine, phenyloin, pilsicainide, propafenone, flecainide, atenolol, acebutolol, sotalol, propranolol, metoprolol, pindolol, amiodarone, nifekalant, diltiazem, bepridil, and verapamil.
  • antihyperlipidemic drug examples include atorvastatin, simvastatin, pravastatin sodium, fluvastatin sodium, clinofibrate, clofibrate, simfibrate, fenofibrate, bezafibrate, colestimide, and colestyramine.
  • immunosuppressant examples include azathioprine, mizoribine, cyclosporine, tacrolimus, gusperimus, and methotrexate.
  • Formulations of the invention may be administered to subjects who have recently received or are likely to receive a dose of radiation or toxin.
  • the dose of radiation or toxin is received as part of a work-related or medical procedure, e.g., working in a nuclear power plant, flying an airplane, an X- ray, CAT scan, or the administration of a radioactive dye for medical imaging; in such an embodiment, the formulation of the invention is administered as a prophylactic measure.
  • the radiation or toxin exposure is received unintentionally, e.g., as a result of an industrial accident, habitation in a location of natural radiation, terrorist act, or act of war involving radioactive or toxic material.
  • the formulation of the invention is preferably administered as soon as possible after the exposure to inhibit apoptosis and the subsequent development of acute radiation syndrome.
  • Formulations of the invention may also be used for treating and/or preventing cancer.
  • formulations of the invention may be used for treating and/or preventing cancer.
  • Calorie restriction has been linked to a reduction in the incidence of age-related disorders including cancer (see e.g., Bordone and Guarente, Nat. Rev. MoI. Cell Biol. (2005 epub); Guarente and Picard, Cell 120: 473-82 (2005); Berrigan, et al., Carcinogenesis 23: 817-822 (2002); and Heilbronn and Ravussin, Am. J. Clin. Nutr.
  • the Sir2 protein from yeast has been- shown to be required for lifespan extension by glucose restriction (see e.g., Lin et al., Science 289: 2126-2128 (2000); Anderson et al., Nature 423: 181 -185 (2003)), a yeast model for calorie restriction. Accordingly, an increase in the level and/or activity of a sirtuin protein may be useful for treating and/or preventing the incidence of age-related disorders, such as, for example, cancer.
  • Exemplary cancers that may be treated using a formulation of the invention are those of the brain and kidney; hormone-dependent cancers including breast, prostate, testicular, and ovarian cancers; lymphomas, and leukemias.
  • a formulation of the invention may be administered directly into the tumor.
  • Cancer of blood cells e.g., leukemia
  • Benign cell growth can also be treated, e.g., warts.
  • Other diseases that can be treated include autoimmune diseases, e.g., systemic lupus erythematosus, scleroderma, and arthritis, in which autoimmune cells should be removed.
  • Viral infections such as herpes, HIV, adenovirus, and HTLV-I associated malignant and benign disorders can also be treated by administration of a formulation of the invention.
  • cells can be obtained from a subject, treated ex vivo to remove certain undesirable cells, e.g., cancer cells, and administered back to the same or a different subject.
  • chemotherapeutic agents may be categorized by their mechanism of action into, for example, following groups: anti-metabolites/anti-cancer agents, such as pyrimidine analogs (5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine) and purine analogs, folate antagonists and related inhibitors (mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine (cladribine)); antiproliferative/antimitotic agents including natural products such as vinca alkaloids (vinblastine, vincristine, and vinorelbine), microtubule disrupters such as taxane (paclitaxel, docetaxel), vincristin, vinblastin, nocodazole, epothilones and navelbine, epidipodophyllotoxins (teniposide), DNA damaging agents (actinomycin, amsacrine, anthracyclines, bleomycin, busulfan,
  • antibiotics such as dactinomycin (actinomycin D), daunorubicin, doxorubicin (adriamycin), idarubicin, anthracyclines, mitoxantrone, bleomycins, plicamycin (mithramycin) and mitomycin; enzymes (L-asparaginase which systemically metabolizes L-asparagine and deprives cells which do not have the capacity to synthesize their own asparagine); antiplatelet agents; antiproliferative/antimitotic alkylating agents such as nitrogen mustards (mechlorethamine, cyclophosphamide and analogs, melphalan, chlorambucil), ethylenimines and methylmelamines (hexamethylmelamine and thiotepa), alkyl sulfonates-busulfan, nitrosoureas (carmus
  • chemotherapeutic agents may be used by themselves with a formulation of the invention.
  • Many combinatorial therapies have been developed, including but not limited to those listed in Table 1.
  • Table 1 Exemplary combinatorial therapies for the treatment of cancer.
  • a formulation of the invention can also be used with antisense RNA, RNAi or other polynucleotides to inhibit the expression of the cellular components that contribute to unwanted cellular proliferation that are targets of conventional chemotherapy.
  • targets are, merely to illustrate, growth factors, growth factor receptors, cell cycle regulatory proteins, transcription factors, or signal transduction kinases.
  • Combination therapies comprising a formulation of the invention and a conventional chemotherapeutic agent may be advantageous over combination therapies known in the art because the combination allows the conventional chemotherapeutic agent to exert greater effect at lower dosage.
  • the effective dose (ED 50 ) for a chemotherapeutic agent, or combination of conventional chemotherapeutic agents, when used in combination with a formulation of the invention is at least 2 fold less than the ED 50 for the chemotherapeutic agent alone, and even more preferably at 5 fold, 10 fold or even 25 fold less.
  • the therapeutic index (TI) for such chemotherapeutic agent or combination of such chemotherapeutic agent when used in combination with a formulation of the invention described herein can be at least 2 fold greater than the TI for conventional chemotherapeutic regimen alone, and even more preferably at 5 fold, 10 fold or even 25 fold greater.
  • formulations of the invention can be used to treat patients suffering from neurodegenerative diseases, and traumatic or mechanical injury to the central nervous system (CNS), spinal cord or peripheral nervous system (PNS).
  • CNS central nervous system
  • PNS peripheral nervous system
  • Neurodegenerative disease typically involves reductions in the mass and volume of the human brain, which may be due to the atrophy and/or death of brain cells, which are far more profound than those in a healthy person that are attributable to aging.
  • Neurodegenerative diseases can evolve gradually, after a long period of normal brain function, due to progressive degeneration (e.g., nerve cell dysfunction and death) of specific brain regions.
  • neurodegenerative diseases can have a quick onset, such as those associated with trauma or toxins. The actual onset of brain degeneration may precede clinical expression by many years.
  • neurodegenerative diseases include, but are not limited to, Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS; Lou Gehrig's disease), diffuse Lewy body disease, chorea- acanthocytosis, primary lateral sclerosis, ocular diseases (ocular neuritis), chemotherapy-induced neuropathies (e.g., from vincristine, paclitaxel, bortezomib), diabetes-induced neuropathies and Friedreich's ataxia.
  • a formulation of the invention can be used to treat these disorders and others as described below.
  • AD is a chronic, incurable, and unstoppable CNS disorder that occurs gradually, resulting in memory loss, unusual behavior, personality changes, and a decline in thinking abilities. These losses are related to the death of specific types of brain cells and the breakdown of connections and their supporting network (e.g. glial cells) between them. AD has been described as childhood development in reverse. In most people with AD, symptoms appear after the age 60. The earliest symptoms include loss of recent memory, faulty judgment, and changes in personality. Later in the disease, those with AD may forget how to do simple tasks like washing their hands. Eventually people with AD lose all reasoning abilities and become dependent on other people for their everyday care. Finally, the disease becomes so debilitating that patients are bedridden and typically develop coexisting illnesses.
  • PD is a chronic, incurable, and unstoppable CNS disorder that occurs ⁇ gradually and results in uncontrolled body movements, rigidity, tremor, and dyskinesia.
  • These motor system problems are related to the death of brain cells in an area of the brain that produces dopamine, a chemical that helps control muscle activity.
  • symptoms appear after age 50.
  • the initial symptoms of PD are a pronounced tremor affecting the extremities,- notably in the hands or lips.
  • Subsequent characteristic symptoms of PD are stiffness or slowness of movement, a shuffling walk, stooped posture, and impaired balance.
  • secondary symptoms such as memory loss, dementia, depression, emotional changes, swallowing difficulties, abnormal speech, sexual dysfunction, and bladder and bowel problems. These symptoms will begin to interfere with routine activities, such as holding a fork or reading a newspaper.
  • people with PD become so profoundly disabled that they are bedridden.
  • ALS motor neuron disease
  • ALS motor neuron disease
  • the motor neurons deteriorate and eventually die, and though a person ' s brain normally remains fully functioning and alert, the command to move never reaches the muscles.
  • Most people who get ALS are between 40 and 70 years old.
  • the first motor neurons that weaken are those controlling the arms or legs. Those with ALS may have trouble walking, they may drop things, fall, slur their speech, and laugh or cry uncontrollably. Eventually the muscles in the limbs begin to atrophy from disuse. This muscle weakness will become debilitating and a person will need a wheel chair or become unable to function out of bed.
  • HD is another neurodegenerative disease resulting from genetically programmed degeneration of neurons in certain areas of the brain. This degeneration causes uncontrolled movements, loss of intellectual faculties, and emotional disturbance.
  • HD is a familial disease, passed from parent to child through a dominant mutation in the wild-type gene. Some early symptoms of HD are mood swings, depression, irritability or trouble driving, learning new things, remembering a fact, or making a decision. As the disease progresses, concentration on intellectual tasks becomes increasingly difficult and the patient may have difficulty feeding himself or herself and swallowing.
  • Tay-Sachs disease and Sandhoff disease are glycolipid storage diseases caused by the lack of lysosomal ⁇ -hexosaminidase (Gravel et al., in The Metabolic Basis of Inherited Disease, eds. Scriver et al., McGraw-Hill, New York, pp. 2839- 2879, 1995).
  • GM2 ganglioside and related glycolipidssubstrates for ⁇ -hexosaminidase accumulate in the nervous system and trigger acute neurodegeneration.
  • the onset of symptoms begins in early infancy.
  • a precipitous neurodegenerative course then ensues, with affected infants exhibiting motor dysfunction, seizure, visual loss, and deafness. Death usually occurs by 2-5 years of age. Neuronal loss through an apoptotic mechanism has been demonstrated (Huang et al., Hum. MoI. Genet. 6: 1879- 1885, 1997).
  • HIV peripheral neuropathies associated with HIV, namely sensory neuropathy, AIDP/CIPD, drug-induced neuropathy and CMV-related.
  • DSPN distal symmetrical polyneuropathy
  • AIDP/CIDP acute or chronic inflammatory demyelinating polyneuropathy
  • AIDP/CIDP there is damage to the fatty membrane covering the nerve impulses.
  • This kind of neuropathy involves inflammation and resembles the muscle deterioration often identified with long-term use of AZT. It can be the first manifestation of HIV infection, where the patient may not complain of pain, but fails to respond to standard reflex tests.
  • This kind of neuropathy may be associated with seroconversion, in which case it can sometimes resolve spontaneously. It can serve as a sign of HIV infection and indicate that it might be time to consider antiviral therapy.
  • AIDP/CIDP may be auto-immune in origin.
  • Drug-induced, or toxic, neuropathies can be very painful. Antiviral drugs commonly cause peripheral neuropathy, as do other drugs e.g. vincristine, dilantin (an anti-seizure medication), high-dose vitamins, isoniazid, and folic acid antagonists. Peripheral neuropathy is often used in clinical trials for antivirals as a dose-limiting side effect, which means that more drugs should not be administered. Additionally, the use of such drugs can exacerbate otherwise minor neuropathies. Usually, these drug-induced neuropathies are reversible with the discontinuation of the drug.
  • CMV causes several neurological syndromes in AIDS, including encephalitis, myelitis, and polyradiculopathy.
  • Neuronal loss is also a salient feature of prion diseases, such as Creutzfeldt- Jakob disease in human, BSE in cattle (mad cow disease), Scrapie Disease in sheep and goats, and feline spongiform encephalopathy (FSE) in cats.
  • Formulations of the invention may be useful for treating or preventing neuronal loss due to these prior diseases.
  • a formulation of the invention may be used to treat or prevent any disease or disorder involving axonopathy.
  • Distal axonopathy is a type of peripheral neuropathy that results from some metabolic or toxic derangement of peripheral nervous system (PNS) neurons. It is the most common response of nerves to metabolic or toxic disturbances, and as such may be caused by metabolic diseases • such as diabetes, renal failure, deficiency syndromes such as malnutrition and alcoholism, or the effects of toxins or drugs.
  • the most common cause of distal axonopathy is diabetes, and the most common distal axonopathy is diabetic neuropathy.
  • the most distal portions of axons are usually the first to degenerate, and axonal atrophy advances slowly towards the nerve's cell body.
  • Diabetic neuropathies are neuropathic disorders that are associated with diabetes mellitus. These conditions usually result from diabetic microvascular injury involving small blood vessels that supply nerves (vasa nervorum). Relatively common conditions which may be associated with diabetic neuropathy include third nerve palsy; mononeuropathy; mononeuritis multiplex; diabetic amyotrophy; a painful polyneuropathy; autonomic neuropathy; and thoracoabdominal neuropathy.
  • diabetic neuropathy Clinical manifestations of diabetic neuropathy include, for example, sensorimotor polyneuropathy such as numbness, sensory loss, dysesthesia and nighttime pain; autonomic neuropathy such as delayed gastric emptying or gastroparesis; and cranial neuropathy such as oculomotor (3rd) neuropathies or Mononeuropathies of the thoracic or lumbar spinal nerves.
  • Peripheral neuropathy is the medical term for damage to nerves of the peripheral nervous system, which may be caused either by diseases of the nerve or from the side-effects of systemic illness. Peripheral neuropathies vary in their presentation and origin, and may affect the nerve or the neuromuscular junction. Major causes of peripheral neuropathy include seizures, nutritional deficiencies, and HIV, though diabetes is the most likely cause.
  • a formulation of the invention may be used to treat or prevent multiple sclerosis (MS), including relapsing MS and monosymptomatic MS, and other demyelinating conditions, such as, for example, chromic inflammatory demyelinating polyneuropathy (CIDP), or symptoms associated therewith.
  • MS is a chronic, often disabling disease of the central nervous system.
  • MS is now a common and well-known neurological disorder that is characterized by episodic patches of inflammation and demyelination which can occur anywhere in the CNS.
  • Demyelination produces a situation analogous to that resulting from cracks or tears in an insulator surrounding an - electrical cord. That is, when the insulating sheath is disrupted, the circuit is "short circuited" and the electrical apparatus associated therewith will function intermittently or nor at all.
  • Such loss of myelin surrounding nerve fibers results in short circuits in nerves traversing the brain and the spinal cord that thereby result in symptoms of MS.
  • demyelination occurs in patches, as opposed to along the entire CNS.
  • demyelination may be intermittent. Therefore, such plaques are disseminated in both time and space. It is believed that the pathogenesis involves a local disruption of the blood brain barrier which causes a localized immune and inflammatory response, with consequent damage to myelin and hence to neurons.
  • MS exists in both sexes and can occur at any age. However, its most common presentation is in the relatively young adult, often with a single focal lesion such as a damage of the optic nerve, an area of anesthesia (loss of sensation), or paraesthesia (localize loss of feeling), or muscular weakness.
  • a single focal lesion such as a damage of the optic nerve, an area of anesthesia (loss of sensation), or paraesthesia (localize loss of feeling), or muscular weakness.
  • vertigo, double vision, localized pain, incontinence, and pain in the amis and legs may occur upon flexing of the neck, as well as a large variety of less common symptoms.
  • An initial attack of MS is often transient, and it may be weeks, months, or years before a further attack occurs.
  • Some individuals may enjoy a stable, relatively event free condition for a great number of years, while other less fortunate ones may experience a continual downhill course ending in complete paralysis.
  • elevated body temperature i.e., a fever, will make the condition worse, or as a reduction of temperature by, for example, a cold bath, may make the condition better.
  • a formulation of the invention may be used to treat trauma to the nerves, including, trauma due to disease, injury (including surgical intervention), or environmental trauma (e.g., neurotoxins, alcoholism, etc.).
  • Formulations of the invention may also be useful to prevent, treat, and alleviate symptoms of various PNS disorders, such as the ones described below.
  • the PNS is composed of the nerves that lead to or branch off from the spinal cord and CNS.
  • the peripheral nerves handle a diverse array of functions in the body, including sensory, motor, and autonomic functions.
  • nerves of the PNS have been damaged. Nerve damage can arise from a number of causes, such as disease, physical injury, poisoning, or malnutrition. These agents may affect either afferent or efferent nerves. Depending on the cause of damage, the nerve cell axon, its protective myelin sheath, or both may be injured or destroyed.
  • peripheral neuropathy encompasses a wide range of disorders in which the nerves outside of the brain and spinal cord — peripheral nerves — have been damaged. Peripheral neuropathy may also be referred to as peripheral neuritis, or if many nerves are involved, the terms polyneuropathy or polyneuritis may be used.
  • Peripheral neuropathy is a widespread disorder, and there are many underlying causes. Some of these causes are common, such as diabetes, and others are extremely rare, such as acrylamide poisoning and certain inherited disorders.
  • the most common worldwide cause of peripheral neuropathy is leprosy. Leprosy is caused by the bacterium Mycobacterium leprae, which attacks the peripheral nerves of affected people. Leprosy is extremely rare in the United States, where diabetes is the most commonly known cause of peripheral neuropathy. It has been estimated that more than 17 million people in the United States and Europe have diabetes-related polyneuropathy. Many neuropathies are idiopathic; no known cause can be found.
  • peripheral neuropathies The most common of the inherited peripheral neuropathies in the United States is Charcot-Marie-Tooth disease, which affects approximately 125,000 persons. Another of the better known peripheral neuropathies is Guillain-Barre syndrome, which arises from complications associated with viral illnesses, such as cytomegalovirus, Epstein-Barr virus, and human immunodeficiency virus (HIV), or bacterial infection, including Campylobacter jejuni and Lyme disease. The worldwide incidence rate is approximately 1.7 cases per 100,000 people annually. Other well-known causes of peripheral neuropathies include chronic alcoholism, infection of the varicella-zoster virus, botulism, and poliomyelitis. Peripheral neuropathy may develop as a primary symptom, or it may be due to another disease.
  • peripheral neuropathy is only one symptom of diseases such as amyloid neuropathy, certain cancers, or inherited neurologic disorders. Such diseases may affect the PNS and the CNS, as well as other body tissues.
  • PNS diseases treatable with formulations of the invention include: Brachial Plexus Neuropathies (diseases of the cervical and first thoracic roots, nerve trunks, cords, and peripheral nerve components of the brachial plexus. Clinical manifestations include regional pain, paresthesia; muscle weakness, and decreased sensation in the upper extremity. These disorders may be associated with trauma, including birth injuries; thoracic outlet syndrome; neoplasms, neuritis, radiotherapy; and other conditions.
  • Diabetic Neuropathies peripheral, autonomic, and cranial nerve disorders that are associated with diabetes mellitus. These conditions usually result from diabetic microvascular injury involving small blood vessels that supply nerves (vasa nervorum).
  • Relatively common conditions which may be associated with diabetic neuropathy include third nerve palsy; mononeuropathy; mononeuritis multiplex; diabetic amyotrophy; a painful polyneuropathy-; autonomic neuropathy; and thoracoabdominal neuropathy (see Adams et al., Principles of Neurology, 6th ed, pi 325); mononeuropathies (disease or trauma involving a single peripheral nerve in isolation, or out of proportion to evidence of diffuse peripheral nerve dysfunction).
  • Mononeuritis multiplex refers to a condition characterized by multiple isolated nerve injuries.
  • Mononeuropathies may result from a wide variety of causes, including ischemia; traumatic injury; compression; connective tissue diseases; cumulative trauma disorders; and other conditions; Neuralgia (intense or aching pain that occurs along the course or distribution of a peripheral or cranial nerve); Peripheral Nervous System Neoplasms (neoplasms which arise from peripheral nerve tissue). This includes neurofibromas; Schwannomas; granular cell tumors; and malignant peripheral nerve sheath tumors (see DeVita Jr et al., Cancer: Principles and Practice of Oncology, 5th ed, ppl 750-1 ); and Nerve Compression Syndromes (mechanical compression of nerves or nerve roots from internal or external causes).
  • causes including ischemia; traumatic injury; compression; connective tissue diseases; cumulative trauma disorders; and other conditions; Neuralgia (intense or aching pain that occurs along the course or distribution of a peripheral or cranial nerve); Peripheral Nervous System Neoplasms (neoplasms
  • nerve impulses due to, for example, myelin sheath dysfunction, or axonal loss.
  • the nerve and nerve sheath injuries may be caused by ischemia; inflammation; or a direct mechanical effect; Neuritis (a general term indicating inflammation of a peripheral or cranial nerve).
  • Clinical manifestation may include pain; paresthesias; paresis; or hyperesthesia; Polyneuropathies (diseases of multiple peripheral nerves).
  • the various forms are categorized by the type of nerve affected (e.g., sensory, motor, or autonomic), by the distribution of nerve injury (e.g., distal vs. proximal), by nerve component primarily affected (e.g., demyelinating vs. axonal), by etiology, or by pattern of inheritance.
  • a formulation of the invention may be used to treat or prevent chemotherapeutic induced neuropathy.
  • the formulation of the invention may be administered prior to administration of the chemotherapeutic agent, concurrently with administration of the chemotherapeutic drug, and/or after initiation of administration of the chemotherapeutic drug. If the formulation of the invention is administered after the initiation of administration of the chemotherapeutic drug, it is desirable that the formulation of the invention be administered prior to, or at the first signs, of chemotherapeutic induced neuropathy.
  • Chemotherapy drugs can damage any part of the nervous system. Encephalopathy and myelopathy are notably very rare. Damage to peripheral nerves is much more common and can be a side effect of treatment experienced by people with cancers, such as lymphoma. Most of the neuropathy affects sensory rather than motor nerves. Thus, the common symptoms are tingling, numbness or a loss of balance. The longest nerves in the body seem to be most sensitive hence the • fact that most patients will report numbness or pins and needles in their hands and feet.
  • the chemotherapy drugs which are most commonly associated with neuropathy are the Vinca alkaloids (anti-cancer drugs originally derived from a member of the periwinkle - the Vinca plant genus) and a platinum- containing drug called Cisplatin.
  • the Vinca alkaloids include the drugs vinblastine, vincristine and vindesine.
  • Many combination chemotherapy treatments for lymphoma for example CHOP and CVP contain vincristine, which is the drug known to cause this problem most frequently. Indeed, it is the risk of neuropathy that limits the dose of vincristine that can be administered.
  • a formulation of the invention may be used to treat or prevent a polyglutamine disease.
  • Huntington's Disease (HD) and Spinocerebellar ataxia type 1 (SCAl) are just two examples of a class of genetic diseases caused by dynamic mutations involving the expansion of triplet sequence repeats. In reference to this common mechanism, these disorders are called trinucleotide repeat diseases. At least 14 such diseases are known to affect human beings. Nine of them, including SCAl and Huntington's disease, have CAG as the repeated sequence (see Table 2 below). Since CAG codes for an amino acid called glutamine, these nine trinucleotide repeat disorders are collectively known as polyglutamine diseases.
  • mice Regardless of whether the mice express full-length proteins or only those portions of the proteins containing the polyglutamine tracts, they develop symptoms of polyglutamine diseases.
  • a long polyglutamine tract by itself is damaging to cells and does not have to be part of a functional protein to cause its damage.
  • SCAl the symptoms of SCAl are not directly caused by the loss of normal ataxin-1 function but rather by the interaction between ataxin- 1 and another protein called LANP.
  • LANP is needed for nerve cells to communicate with one another and thus for their survival.
  • the mutant ataxin-1 protein accumulates inside nerve cells, it "traps'- the LANP protein, interfering with its normal function. After a while, the absence of LANP function appears to cause nerve cells to malfunction.
  • HDAC I/II Class I/II Histone Deacetylase
  • the invention provides a method for treating or preventing neuropathy related to ischemic injuries or diseases, such as, for example, coronary heart disease (including congestive heart failure and myocardial infarctions), stroke, emphysema, hemorrhagic shock, peripheral vascular disease (upper and lower extremities) and transplant related injuries.
  • ischemic injuries or diseases such as, for example, coronary heart disease (including congestive heart failure and myocardial infarctions), stroke, emphysema, hemorrhagic shock, peripheral vascular disease (upper and lower extremities) and transplant related injuries.
  • the invention provides a method to treat a central nervous system cell to prevent damage in response to a decrease in blood flow to the cell.
  • the severity of damage that may be prevented will depend in large part on the degree of reduction in blood flow to the cell and the duration of the reduction.
  • the normal amount of perfusion to brain gray matter in humans is about 60 to 70 mL/100 g of brain tissue/min.
  • Death of central nervous system cells typically occurs when the flow of blood falls below approximately 8-10 mL/100 g of brain tissue/min, while at slightly higher levels (i.e. 20-35 mL/100 g of brain tissue/min) the tissue remains alive but not able to function.
  • apoptotic or necrotic cell death may be prevented.
  • ischemic-mediated damage such as cytoxic edema or central nervous system tissue anoxemia, may be prevented.
  • the central nervous system cell may be a spinal cell or a brain cell.
  • the ischemic condition is a stroke that results in any type of ischemic central nervous system damage, such as apoptotic or necrotic cell death, cytoxic edema or central nervous system tissue anoxia.
  • the stroke may impact any area of the brain or be caused by any etiology commonly known to result in the occurrence of a stroke.
  • the stroke is a brain stem stroke.
  • brain stem strokes strike the brain stem, which control involuntary life-support functions such as breathing, blood pressure, and heartbeat.
  • the stroke is a cerebellar stroke.
  • cerebellar strokes impact the cerebellum area of the brain, which controls balance and coordination.
  • the stroke is an embolic stroke.
  • embolic strokes may impact any region of the brain and typically result from the blockage of an artery by a vaso-occlusion.
  • the stroke may be a hemorrhagic stroke.
  • hemorrhagic stroke may impact any region of the brain, and typically result from a ruptured blood vessel characterized by a hemorrhage (bleeding) within or surrounding the brain.
  • the stroke is a thrombotic stroke.
  • thrombotic strokes result from the blockage of a blood vessel by accumulated deposits.
  • the ischemic condition may result from a disorder that occurs in a part of the subject's body outside of the central nervous system, but yet still causes a reduction in blood flow to the central nervous system.
  • disorders may include, but are not limited to a peripheral vascular disorder, a venous thrombosis, a pulmonary embolus, arrhythmia (e.g. atrial fibrillation), a myocardial infarction, a transient ischemic attack, unstable angina, or sickle cell anemia.
  • the central nervous system ischemic condition may occur as result of the subject undergoing a surgical procedure.
  • the subject may be undergoing heart surgery, lung surgery, spinal surgery; brain surgery, vascular surgery, abdominal surgery, or organ transplantation surgery.
  • the organ transplantation surgery may include heart, lung, pancreas, kidney or liver transplantation surgery.
  • the central nervous system ischemic condition may occur as a result of a trauma or injury to a part of the subject's body outside the central nervous system.
  • the trauma or injury may cause a degree of bleeding that significantly reduces the total volume of blood in the subject's body. Because of this reduced total volume, the amount of blood flow to the central nervous system is concomitantly reduced.
  • the trauma or injury may also result in the formation of a vaso-occlusion that restricts blood flow to the central nervous system.
  • the formulations of the invention may be employed to treat the central nervous system ischemic condition irrespective of the cause of the condition.
  • the ischemic condition results from a vaso-occlusion.
  • the vaso-occlusion may be any type of occlusion, but is typically a cerebral thrombosis or an embolism.
  • the ischemic condition may result from a hemorrhage.
  • the hemorrhage may be any type of hemorrhage, but is generally a cerebral hemorrhage or a subararachnoid hemorrhage.
  • the ischemic condition may result from the narrowing of a vessel. Generally speaking, the vessel may narrow as a result of a vasoconstriction such as occurs during vasospasms, or due to arteriosclerosis.
  • the ischemic condition results from an injury to the brain or spinal cord.
  • a formulation of the invention may be administered to reduce infarct size of the ischemic core following a central nervous system ischemic condition. Moreover, a formulation of the invention may also be beneficially administered to reduce the size of the ischemic penumbra or transitional zone following a central nervous system ischemic condition.
  • a combination drug regimen may include drugs or compounds for the treatment or prevention of neurodegenerative disorders or secondary conditions associated with these conditions.
  • a combination drug regimen may include a formulation of the invention and one or more anti- neurodegeneration agents.
  • a formulation of the invention can be combined with an effective amount of one or more of: L-DOPA; a dopamine agonist; an adenosine A 2 A receptor antagonist; a COMT inhibitor; a MAO inhibitor; an N-NOS inhibitor; a sodium channel antagonist; a selective N -methyl D- aspartate (NMDA) receptor antagonist; an AMPA/kainate receptor antagonist; a calcium channel antagonist; a GABA-A receptor agonist; an acetyl-choline esterase inhibitor; a matrix metalloprotease inhibitor; a PARP inhibitor; an inhibitor of p38 MAP kinase or c-jun-N-terminal kinases; TPA; NDA antagonists; beta-interferons; growth factors; glutamate inhibitors;
  • N-NOS inhibitors include 4-(6-amino-pyridin-2-yl)-3- methoxyphenol 6-[4-(2-dimethylamino-ethoxy)-2-methoxy-phenyl]-pyridin-2-yl- amine, 6-[4-(2-dimethylamino-ethoxy)-2,3-dimet-hyl-phenyl]-pyridin-2-yl-amine, 6-[4-(2-pyrrolidinyl-ethoxy)-2,3-dimethyl-p-henyl]-pyridin-2-yl-amine, 6-[4-(4-(n- methyl)piperidinyloxy)-2,3-dimethyl-p-henyl]-pyridin-2-yl-amine, 6-[4-(2- dimethylamino-ethoxy)-3-methoxy-phenyl]-pyridin-2-yl-amine, 6-[4-(2- pyrrolidinyl-ethoxy
  • Exemplary NMDA receptor antagonists include (+)-( 1 S, 2S)- 1 -(4-hydroxy- phenyl)-2-(4-hydroxy-4-phenylpiperidino)- l -pro-panol, (I S, 2S)-l -(4-hydroxy-3- methoxyphenyl)-2-(4-hydroxy-4-phenylpiperi-dino)-l -propanol, (3R, 4S)-3-(4-(4- fluorophenyl)-4-hydroxypiperidin-l-yl-)-chroman-4,7-diol, (IR*, 2R*)-l -(4- hydroxy-3-methylphenyl)-2-(4-(4-fluoro-phenyl)-4-hydroxypiperidin-l-yl)-propan- 1 -ol-mesylate or a pharmaceutically acceptable acid addition salt thereof.
  • dopamine agonists include ropininole; L-dopa decarboxylase inhibitors such as carbidopa or benserazide, bromocriptine, dihydroergocryptine, etisulergine, AF- 14, alaptide, pergolide, piribedil; dopamine Dl receptor agonists such as A-68939, A-77636, dihydrexine, and SKF-38393; dopamine D2 receptor agonists such as carbergoline, lisuride, N-0434, naxagolide, PD-1 18440, pramipexole, quinpirole and ropinirole; dopamine/ ⁇ -adrenegeric receptor agonists such as DPDMS and dopexamine; dopamine/5-HT uptake inhibitor/5-HT- 1 A agonists such as roxindole; dopamine/opiate receptor agonists such as NIH- 10494; ⁇
  • Exemplary acetyl cholinesterase inhibitors include donepizil, 1 -(2-methyl- lH-benzimida-zol-5-yl)-3-[ l -(phenylmethyl)-4-piperidinyl]-l -propanone; l -(2- phenyl- 1 H-benzimidazol-5-yl)-3-[ 1 -(phenylmethyl)-4-piperidinyl]- 1 -pr-opanone; 1 - ( 1 -ethyl-2-methyl- 1 H-benzimidazol-5-yl)-3 -[ 1 -(phenylmethyl)-4-p-iperidinyl]- 1 - propanone; 1 -(2-methyl-6-benzothiazolyl)-3-[ 1 -(phenylmethyl)-4-piperidinyl]- 1 - propanone; 1 -(2-methyl-6-benzothiazolyl)-3-[ 1 -[(2-methyl-4
  • Exemplary calcium channel antagonists include diltiazem, omega-conotoxin GVIA, methoxyverapamil, amlodipine, felodipine, lacidipine, and mibefradil.
  • GABA-A receptor modulators include clomethiazole; IDDB; gaboxadol (4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol); ganaxolone (3 ⁇ - hydroxy-3 ⁇ -methyl-5 ⁇ -pregnan-20-one); fengabine (2-[(butylimino)-(2- chlorophenyl)methyl]-4-chlorophenol); 2-(4-methoxyphenyl)-2,5,6,7,8,9- hexahydro-pyrazolo[4,3-c]cinnolin-3-one; 7-cyclobutyl-6-(2-methyl-2H-l ,2,4- triazol-3-ylmethoxy)-3-phenyl-l ,2,4-triazolo[4,3-b]pyridazine; (3-fluoro-4- methylphenyl)-N-( ⁇ -l -[(2-methylphenyl)methyl]-
  • Exemplary potassium channel openers include diazoxide, flupirtine, pinacidil, levcromakalim, rilmakalim, chromakalim, PCO-400 and SKP-450 (2-
  • Exemplary AiMPA/kainate receptor antagonists include 6-cyano-7- nitroquinoxalin-2,3-di-one (CNQX); 6-nitro-7-sulphamoylbenzo[fjquinoxaline-2,3- dione (NBQX); 6,7-dinitroquinoxaline-2,3-dione (DNQX); 1 -(4-aminophenyl)-4- methyl-7,8-methylenedioxy-5H-2,3-benzodiazepine hydrochloride; and 2,3- dihydroxy-6-nitro-7-sulfamoylbenzo-[f]quinoxaline.
  • Exemplary sodium channel antagonists include ajmaline, procainamide, flecainide and riluzole.
  • Exemplary matrix-metalloprotease inhibitors include 4-[4-(4- fluorophenoxy)benzenesulfon-ylamino]tetrahydropyran-4-carboxylic acid hydroxyamide; 5-Methyl-5-(4-(4'-fluorophenoxy)-phenoxy)-pyrimidine-2,4,6- trione; 5-n-Butyl-5-(4-(4'-fluorophenoxy)-phenoxy)-pyrimidine-2,4,6-trione and prinomistat.
  • PARP PoIy(ADP ribose) polymerase
  • ADP ribose polymerase PARP
  • PARP is an abundant nuclear enzyme which is activated by DNA strand single breaks to synthesize poly (ADP ribose) from NAD.
  • PARP is involved in base excision repair caused by oxidative stress via the activation and recruitment of DNA repair enzymes in the nucleus.
  • PARP plays a role in cell necrosis and DNA repair.
  • PARP also participates in regulating cytokine expression that mediates inflammation.
  • PARP is over-activated, resulting in cell-based energetic failure characterized by NAD depletion and leading to ATP consumption, cellular necrosis, tissue injury, and organ damage/failure.
  • PARP is thought to contribute to neurodegeneration by depleting nicotinamide adenine dinucleotide (NAD+) which then reduces adenosine triphosphate (ATP; Cosi and Marien, Ann. N. Y. Acad. Sci., 890:227, 1999) contributing to cell death which can be prevented by PARP inhibitors.
  • NAD+ nicotinamide adenine dinucleotide
  • ATP adenosine triphosphate
  • Exemplory PARP inhibitors can be found in Southan and Szabo, Current Medicinal Chemistry, 10:321 , 2003.
  • Exemplary inhibitors of p38 MAP kinase and c-jun-N-te ⁇ ninal kinases include pyridyl imidazoles, such as PD 169316, isomeric PD 1693 16, SB 203580, SB 202190, SB 220026, and RWJ 67657. Others are described in US Patent 6,288,089, and incorporated by reference herein.
  • a combination therapy for treating or preventing MS comprises a formulation of the invention and one or more of Avonex ® (interferon beta- I a), Tysabrr (natalizumab), or Fumaderm ® (BG- 12/Or-al Fumarate).
  • Avonex ® interferon beta- I a
  • Tysabrr natalizumab
  • Fumaderm ® BG- 12/Or-al Fumarate
  • a combination therapy for treating or preventing diabetic neuropathy or conditions associated therewith comprises a therapeutically effective amount of formulation of the invention and one or more of tricyclic antidepressants (TCAs) (including, for example, imipramine, amytriptyline, desipramine and nortriptyline), serotonin reuptake inhibitors (SSRIs) (including, for example, fluoxetine, paroxetine, sertralene, and citalopram) and antiepileptic drugs (AEDs) (including, for example, gabapentin, carbamazepine, and topimirate).
  • TCAs tricyclic antidepressants
  • SSRIs serotonin reuptake inhibitors
  • AEDs antiepileptic drugs
  • the invention provides a method for treating or preventing a polyglutamine disease using a combination comprising formulation of the invention and at least one HDAC I/II inhibitor.
  • HDAC I/II inhibitors include hydroxamic acids, cyclic peptides, benzamides, short-chain fatty acids, and depudecin.
  • hydroxamic acids and hydroxamic acid derivatives include, but are not limited to, trichostatin A (TSA), suberoylanilide hydroxamic acid (SAHA), oxamflatin, suberic bishydroxamic acid (SBHA), m-carboxy-cinnamic acid bishydroxamic acid (CBHA), valproic acid and pyroxamide.
  • TSA was isolated as an antifungi antibiotic (Tsuji et al (1976) J. Antibiot (Tokyo) 29:1 -6) and found to be a potent inhibitor of mammalian HDAC (Yoshida et al. (1990) J. Biol. Chem. 265: 17174-17179).
  • hydroxamic acid-based HDAC inhibitors SAHA, SBHA, and CBHA are synthetic compounds that are able to inhibit HDAC at micromolar concentration or lower in vitro or in vivo. Glick et al. (1999) Cancer Res. 59:4392-4399.
  • SAHA, SBHA, and CBHA are synthetic compounds that are able to inhibit HDAC at micromolar concentration or lower in vitro or in vivo.
  • CBHA hydroxamic acid-based HDAC inhibitors
  • SAHA, SBHA, and CBHA are synthetic compounds that are able to inhibit HDAC at micromolar concentration or lower in vitro or in vivo. Glick et al. (1999) Cancer Res. 59:4392-4399.
  • These hydroxamic acid-based HDAC inhibitors all possess an essential structural feature: a polar hydroxamic terminal linked through a hydrophobic methylene spacer (e.g. 6 carbon at length) to another polar site that is attached to a terminal hydrophobic mo
  • Cyclic peptides used as HDAC inhibitors are mainly cyclic tetrapeptides.
  • cyclic peptides include, but are not limited to, trapoxin A, apicidin and depsipeptide.
  • Trapoxin A is a cyclic tetrapeptide that contains a 2-amino-8-oxo- 9, 10-epoxy-decanoyl (AOE) moiety.
  • AOE 2-amino-8-oxo- 9, 10-epoxy-decanoyl
  • Depsipeptide is isolated from Chromobacterium violaceum, and has been shown to inhibit HDAC activity at micromolar concentrations.
  • benzamides include but are not limited to MS-27-275. Saito et al. (1990) Proc. Natl. Acad. Sci. USA. 96:4592-4597.
  • short-chain fatty acids include but are not limited to butyrates (e.g., butyric acid, arginine butyrate and phenylbutyrate (PB)).
  • PB phenylbutyrate
  • depudecin which has been shown to inhibit HDAC at micromolar concentrations (Kwon et al. (1998) Proc. Natl. Acad. Sci. USA. 95:3356-3361) also falls within the scope of histone deacetylase inhibitors as described herein. Blood Coagulation Disorders
  • formulations of the invention can be used to treat or prevent blood coagulation disorders (or hemostatic disorders).
  • blood coagulation disorders or hemostatic disorders
  • the terms “hemostasis” , “blood coagulation, “ and “blood clotting” refer to the control of bleeding, including the physiological properties of vasoconstriction and coagulation. Blood coagulation assists in maintaining the integrity of mammalian circulation after injury, inflammation, disease, congenital defect, dysfunction or other disruption. After initiation of clotting, blood coagulatron proceeds through the sequential activation of certain plasma proenzymes to their enzyme forms (see, for example, Coleman, R. W. et al. (eds.) Hemostasis and Thrombosis, Second Edition, (1987)).
  • Plasma glycoproteins including Factor XII, Factor XI, Factor IX, Factor X, Factor VII, and prothrombin, are zymogens of serine proteases. Most of these blood clotting enzymes are effective on a physiological scale only when assembled in complexes on membrane surfaces with protein cofactors such as Factor VIII and Factor V. Other blood factors modulate and localize clot formation, or dissolve blood clots.
  • Activated protein C is a specific enzyme that inactivates procoagulant components. Calcium ions are involved in many of the component reactions.
  • Blood coagulation follows either the intrinsic pathway, where all of the protein components are present in blood, or the extrinsic pathway, where the cell-membrane protein tissue factor plays a critical role. Clot formation occurs when fibrinogen is cleaved by thrombin to form fibrin. Blood clots are composed of activated platelets and fibrin.
  • the formation of blood clots does not only limit bleeding in case of an injury (hemostasis), but may lead to serious organ damage and death in the context of atherosclerotic diseases by occlusion of an important artery or vein.
  • Thrombosis is thus blood clot formation at the wrong time and place. It involves a cascade of complicated and regulated biochemical reactions between circulating blood proteins (coagulation factors), blood cells (in particular platelets), and elements of an injured vessel wall.
  • the present invention provides anticoagulation and antithrombotic treatments aiming at inhibiting the formation of blood clots in order to prevent or treat blood coagulation disorders, such as myocardial infarction, stroke, loss of a limb by peripheral artery disease or pulmonary embolism.
  • modulating or modulation of hemostasis includes the induction (e.g., stimulation or increase) of hemostasis, as well as the inhibition (e.g., reduction or decrease) of hemostasis.
  • the invention provides a method for reducing or inhibiting hemostasis in a subject by administering a formulation of the invention.
  • the compositions and methods disclosed herein are useful for the treatment or prevention of thrombotic disorders.
  • thrombotic disorder includes any disorder or condition characterized by excessive or unwanted coagulation or hemostatic activity, or a hypercoagulable state.
  • thrombootic disorders include diseases or disorders involving platelet adhesion and thrombus formation, and may manifest as an increased propensity to form thromboses, e.g., an increased number of thromboses, thrombosis at an early age, a familial tendency towards thrombosis, and thrombosis at unusual sites.
  • thrombotic disorders include, but are not limited to, thromboembolism, deep vein thrombosis, pulmonary embolism, stroke, myocardial infarction, miscarriage, thrombophilia associated with anti-thrombin III deficiency, protein C deficiency, protein S deficiency, resistance to activated protein C, dysfibrinogenemia, fibrinolytic disorders, homocystinuria, pregnancy, inflammatory disorders, myeloproliferative disorders, arteriosclerosis, angina, e.g., unstable angina, disseminated intravascular coagulation, thrombotic thrombocytopenic purpura, cancer metastasis, sickle cell disease, glomerular nephritis, and drug induced thrombocytopenia (including, for example, heparin induced thrombocytopenia).
  • formulation of the inventions may be administered to prevent thrombotic events or to prevent re-occlusion during or after therapeutic intervention
  • a combination drug regimen may include drugs or compounds for the treatment or prevention of blood coagulation disorders or secondary conditions associated with these conditions.
  • a combination drug regimen may include a formulation of the invention and one or more anticoagulation or anti-thrombosis agents.
  • a formulation of the invention can be combined with an effective amount of one or more of: aspirin, heparin, and oral Warfarin that inhibits Vit K-dependent factors, low molecular weight heparins that inhibit factors X and II, thrombin inhibitors, inhibitors of platelet GP HbIIIa receptors, inhibitors of tissue factor (TF), inhibitors of human von Willebrand factor, inhibitors of one or more factors involved in hemostasis (in particular in the coagulation cascade).
  • formulations of the invention can be combined with thrombolytic agents, such as t-PA, streptokinase, reptilase, TNK-t-PA, and staphylokinase.
  • a formulation of the invention may be used for treating or preventing weight gain or obesity in a subject.
  • a formulation of the invention may be used, for example, to treat or prevent hereditary obesity, dietary obesity, hormone related obesity, obesity related to the administration of medication, to reduce the weight of a subject, or to reduce or prevent weight gain in a subject.
  • a subject in need of such a treatment may be a subject who is obese, likely to become obese, overweight, or likely to become overweight-Subjects who are likely to become obese or overweight can be identified, for example, based on family history, genetics, diet, activity level, medication intake, or various combinations thereof.
  • a formulation of the invention may be administered to subjects suffering from a variety of other diseases and conditions that may be treated or prevented by promoting weight loss in the subject.
  • diseases include, for example, high blood pressure, hypertension, high blood cholesterol, dyslipidemia, type 2 diabetes, insulin resistance, glucose intolerance, hyperinsulinemia, coronary heart disease, angina pectoris, congestive heart failure, stroke, gallstones, cholescystitis and cholelithiasis, gout, osteoarthritis, obstructive sleep apnea and respiratory problems, some types of cancer (such as endometrial, breast, prostate, and colon), complications of pregnancy, poor female reproductive health (such as menstrual irregularities, infertility, irregular ovulation), bladder control problems (such as stress incontinence); uric acid nephrolithiasis; psychological disorders (such as depression, eating disorders, distorted body image, and low self esteem). Stunkard AJ, Wadden TA. (Editor
  • a formulation of the invention may be used for inhibiting adipogenesis or fat cell differentiation, whether in vitro or in vivo.
  • high circulating levels of insulin and/or insulin like growth factor (IGF) 1 will be prevented from recruiting preadipocytes to differentiate into adipocytes.
  • IGF insulin like growth factor
  • a formulation of the invention may be used for reducing appetite and/or increasing satiety, thereby causing weight loss or avoidance of weight gain.
  • a subject in need of such a treatment may be a subject who is overweight, obese or a subject likely to become overweight or obese.
  • the method may comprise administering daily or, every other day, or once a week, a dose, e.g., in the form of a pill, to a subject.
  • the dose may be an "appetite reducing dose.'
  • a method for stimulating adipogenesis may comprise contacting a cell with a formulation of the invention.
  • a method for modulating weight may further comprise monitoring the weight of the subject and/or the level of modulation of sirtuins, for example, in adipose tissue.
  • a formulation of the invention may be administered as a combination therapy for treating or preventing weight gain or obesity.
  • a formulation of the invention may be administered in combination with one or more anti-obesity agents.
  • anti-obesity agents include, for example, phenylpropanolamine, ephedrine, pseudoephedrine, phentermine, a cholecystokinin-A agonist, a monoamine reuptake inhibitor (such as sibutramine), a sympathomimetic agent, a serotonergic agent (such as dexfenfluramine or fenfluramine), a dopamine agonist (such as bromocriptine), a melanocyte-stimulating hormone receptor agonist or mimetic, a melanocyte- stimulating hormone analog, a cannabinoid receptor antagonist, a melanin concentrating hormone antagonist, the OB protein (leptin), a leptin analog, a leptin receptor
  • anorectic agents include bombesin agonists, dehydroepiandiOsterone or analogs thereof, glucocorticoid receptor agonists and antagonists, orexin receptor antagonists, urocortin binding protein antagonists, agonists of the glucagon-like peptide- 1 receptor such as Exendin and ciliary neurotrophic factors such as Axokine.
  • a formulation of the invention may be administered to reduce drug-induced weight gain.
  • a formulation of the invention - may be administered as a combination therapy with medications that may stimulate appetite or cause weight gain, in particular, weight gain due to factors other than water retention.
  • Examples of medications that may cause weight gain include for example, diabetes treatments, including, for example, sulfonylureas (such as glipizide and glyburide), thiazolidinediones (such as pioglitazone and rosiglitazone), meglitinides, nateglinide, repaglinide, sulphonylurea medicines, and insulin; antidepressants, including, for example, tricyclic antidepressants (such as amitriptyline and imipramine), irreversible monoamine oxidase inhibitors (MAOIs), selective serotonin reuptake inhibitors (SSRIs), bupropion, paroxetine, and mirtazapine; steroids, such as, for example, prednisone; hormone therapy; lithium carbonate; valproic acid; carbamazepine; chlorpromazine; thiothixene; beta blockers (such as propranolol); alpha blockers (such as
  • formulations of the invention may be used for treating or preventing a metabolic disorder, such as insulin-resistance, a pre-diabetic state, type II diabetes, and/or complications thereof.
  • Administration of a formulation of the invention may increase insulin sensitivity and/or decrease insulin levels in a subject.
  • a subject in need of such a treatment may be a subject who has insulin resistance or other precursor symptom of type II diabetes, who has type II diabetes, or who is likely to develop any of these conditions.
  • the subject may be a subject having insulin resistance, e.g., having high circulating levels of insulin and/or associated conditions, such as hyperlipidemia, dyslipogenesis, hypercholesterolemia, impaired glucose tolerance, high blood glucose sugar level, other manifestations of syndrome X, hypertension, atherosclerosis and lipodystrophy.
  • insulin resistance e.g., having high circulating levels of insulin and/or associated conditions, such as hyperlipidemia, dyslipogenesis, hypercholesterolemia, impaired glucose tolerance, high blood glucose sugar level, other manifestations of syndrome X, hypertension, atherosclerosis and lipodystrophy.
  • formulations of the invention may be administered as a combination therapy for treating or preventing a metabolic disorder.
  • a formulation of the invention may be administered in combination with one or more anti-diabetic agents.
  • anti-diabetic agents include, for example, an aldose reductase inhibitor, a glycogen phosphorylase inhibitor, a sorbitol dehydrogenase inhibitor, a protein tyrosine phosphatase 1 B inhibitor, a dipeptidyl protease inhibitor, insulin (including orally bioavailable insulin preparations), an insulin mimetic, metformin, acarbose, a peroxisome proliferator-activated receptor- ⁇ (PPAR- ⁇ ) ligand such as troglitazone, rosaglitazone, pioglitazone or GW- 1929, a sulfonylurea, glipazide, glyburide, or - chlorpropamide wherein the amounts of the first and second compounds
  • anti-diabetic agents include a glucosidase inhibitor, a glucagon-like peptide- 1 (GLP-I ), insulin, a PPAR ⁇ / ⁇ dual agonist, a meglitimide and an ⁇ P2 inhibitor.
  • an anti-diabetic agent may be a dipeptidyl peptidase IV (DP-IV or DPP-IV) inhibitor, such as, for example LAF237 from Novartis (NVP DPP728; l -[[[2-[(5-cyanopyridin-2-yl)amino] ethyl]amino]acetyl]-2- cyano-(S)- pyrrolidine) or MK-04301 from Merck (see e.g., Hughes et al., Biochemistry 38: 1 1597-603 (1999)). Inflammatory Diseases
  • DP-IV or DPP-IV dipeptidyl peptidase IV
  • formulations of the invention can be used to treat or prevent a disease or disorder associated with inflammation.
  • Formulations of the invention may be administered prior to the onset of, at, or after the initiation of inflammation.
  • the formulations of the invention are preferably provided in advance of any inflammatory response or symptom.
  • Administration of the compounds may prevent or attenuate inflammatory responses or symptoms.
  • Exemplary inflammatory conditions include, for example, multiple sclerosis, rheumatoid arthritis, psoriatic arthritis, degenerative joint disease, spondouloarthropathies, gouty arthritis, systemic lupus erythematosus, juvenile arthritis, rheumatoid arthritis, osteoarthritis, osteoporosis, diabetes (e.g., insulin dependent diabetes mellitus or juvenile onset diabetes), menstrual cramps, cystic fibrosis, inflammatory bowel disease, irritable bowel syndrome, Crohn's disease, mucous colitis, ulcerative colitis, gastritis, esophagitis, pancreatitis, peritonitis, Alzheimer's disease, shock, ankylosing spondylitis, gastritis, conjunctivitis, pancreatis (acute or chronic), multiple organ injury syndrome (e.g., secondary to septicemia or trauma), myocardial infarction, atherosclerosis, stroke, reperfusion
  • Exemplary inflammatory conditions of the skin include, for example, eczema, atopic dermatitis, contact dermatitis, urticaria, scleroderma, psoriasis, and dermatosis with acute inflammatory components.
  • a formulation of the invention may be used to treat or prevent allergies and respiratory conditions, including asthma, bronchitis, pulmonary fibrosis, allergic rhinitis, oxygen toxicity, emphysema, chronic bronchitis, acute respiratory distress syndrome, and any chronic obstructive pulmonary disease (COPD).
  • the formulation of the invention may be used to treat chronic hepatitis infection, including hepatitis B and hepatitis C.
  • a formulation of the invention may be used to treat autoimmune diseases and/or inflammation associated with autoimmune diseases such as organ-tissue autoimmune diseases (e.g., Raynaud's syndrome), scleroderma, myasthenia gravis, transplant rejection, endotoxin shock, sepsis, - psoriasis, eczema, dermatitis, multiple sclerosis, autoimmune thyroiditis, uveitis, systemic lupus erythematosis, Addison's disease, autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome), and Grave's disease.
  • organ-tissue autoimmune diseases e.g., Raynaud's syndrome
  • scleroderma myasthenia gravis
  • transplant rejection transplant rejection
  • endotoxin shock sepsis
  • - psoriasis eczema
  • dermatitis e.g., multiple sclerosis
  • a formulation of the invention may be taken alone or in combination with other compounds useful for treating or preventing inflammation.
  • anti-inflammatory agents include, for example, steroids (e.g., Cortisol, cortisone, fludrocortisone, prednisone, 6 ⁇ -methylprednisone, triamcinolone, betamethasone or dexamethasone), nonsteroidal antiinflammatory drugs (NSAIDS (e.g., aspirin, acetaminophen, tolmetin, ibuprofen, mefenamic acid, piroxicam, nabumetone, rofecoxib, celecoxib, etodolac or nimesulide).
  • steroids e.g., Cortisol, cortisone, fludrocortisone, prednisone, 6 ⁇ -methylprednisone, triamcinolone, betamethasone or dexamethasone
  • NSAIDS nonsteroidal antiinflammatory drugs
  • the other therapeutic agent is an antibiotic (e.g., vancomycin, penicillin, amoxicillin, ampicillin, cefotaxime, ceftriaxone, cefixime, rifampinmetronidazole, doxycycline or streptomycin).
  • the other therapeutic agent is a PDE4 inhibitor (e.g., roflumilast or rolipram).
  • the other therapeutic agent is an antihistamine (e.g., cyclizine, hydroxyzine, promethazine or diphenhydramine).
  • the other therapeutic agent is an anti-malarial (e.g., artemisinin, artemether, artsunate, chloroquine phosphate, mefloquine hydrochloride, doxycycline hyclate, proguanil hydrochloride, atovaquone or halofantrine).
  • the other therapeutic agent is drotrecogin alfa.
  • anti-inflammatory agents include, for example, aceclofenac, acemetacin, e-acetamidocaproic acid, acetaminophen, acetaminosalol, acetanilide, acetylsalicylic acid, S-adenosylmethioninc, alclofenac, alclometasone, alfentanil, algestone, allylprodine, alminoprofen; aloxiprin, alphaprodine, aluminum bis(acetylsalicylate), amcinonide, amfenac, aminochlorthenoxazin, 3- amino-4-hydroxybutyric acid, -2-amino-4-picoline, aminopropylon, aminopyrine, amixetrine, ammonium salicylate, ampiroxicam, amtolmetin guacil, anileridine, antipyrine, antrafenine, apazone, beclomethasone, bendazac, benorylate, benox
  • a formulation of the invention may be administered with a selective COX-2 inhibitor for treating or preventing inflammation.
  • selective COX-2 inhibitors include, for example, deracoxib, parecoxib, celecoxib, valdecoxib, rofecoxib, etoricoxib, lumiracoxib, 2- (3,5-difluorophenyl)-3-[4-(methylsulfonyl)phenyl]-2-cyclopenten-l -one, (S)-6,8- dichloro-2-(triflu- oromethyl)-2H-l -benzopyran-3-carboxylic acid, 2-(3,4- difluorophenyl)-4-(3-hydroxy-3-methyl-l-butoxy)-5-[4-(methylsulfonyl)phenyl]-3- (2H)-pyridazinone, 4-[5-(4-fluorophenyl)-3-(trifluoromethyl)-l H-pyrazol
  • formulations of the invention may be used for reducing the incidence or severity of flushing and/or hot flashes which are symptoms of a disorder.
  • the subject method includes the use of formulations of the invention, alone or in combination with other agents, for reducing incidence or severity of flushing and/or hot flashes in cancer patients.
  • the method provides for the use of formulations of the invention to reduce the incidence or severity of flushing and/or hot flashes in menopausal and post-menopausal woman.
  • formulations of the invention may be used as a therapy for reducing the incidence or severity of flushing and/or hot flashes which are side.- effects of another drug therapy, e.g., drug-induced flushing.
  • a method for treating and/or preventing drug-induced flushing comprises administering to a patient in need thereof a formulation comprising at least one flushing inducing compound and a formulation of the invention.
  • a method for treating drug induced flushing comprises separately administering one or more compounds that induce flushing and a formulation of the invention, e.g., wherein the formulation of the invention and flushing inducing agent have not been formulated in the same compositions.
  • the formulation of the invention may be administered ( 1 ) at the same as administration of the flushing inducing agent, (2) intermittently with the flushing inducing agent, (3) staggered relative to administration of the flushing inducing agent, (4) prior to administration of the flushing inducing agent, (5) subsequent to administration of the flushing inducing agent, and (6) various combination thereof.
  • flushing inducing agents include, for example, niacin, faloxifene, antidepressants, anti-psychotics, chemotherapeutics, calcium channel blockers, and antibiotics.
  • formulations of the invention may be used to reduce flushing side effects of a vasodilator or an antilipemic agent (including anticholesteremic agents and lipotropic agents).
  • a formulation of the invention may be used to reduce flushing associated with the administration of niacin.
  • Nicotinic acid 3-pyridinecarboxylic acid or niacin
  • Nicotinic acid is an antilipidemic agent that is marketed under, for example, the trade names Nicolar ® , SloNiacin ® , Nicobid ® and Time Release Niacin ® .
  • Nicotinic acid has been used for many years in the treatment of lipidemic disorders such as hyperlipidemia, hypercholesterolemia and atherosclerosis. This compound has long been known to exhibit the beneficial effects of reducing total cholesterol, low density lipoproteins or "LDL cholesterol," triglycerides and apolipoprotein a (Lp(a)) in the human body, while increasing desirable high density lipoproteins or "HDL cholesterol".
  • Typical doses range from about 1 gram to about 3 grams daily. Nicotinic acid is normally administered two to four times per day after meals, depending upon the dosage form selected. Nicotinic acid isxurrently commercially available in two dosage forms. One dosage form is an immediate or rapid release tablet which should be administered three or four times per day. Immediate release (“IR”) nicotinic acid formulations generally release nearly all of their nicotinic acid within about 30 to 60 minutes following ingestion. The other dosage form is a sustained release form which is suitable for administration two to four times per day.
  • IR immediate release
  • sustained release (“SR") nicotinic acid formulations are designed to release significant quantities of drug for absorption into the blood stream over specific timed intervals in order to maintain therapeutic levels of nicotinic acid over ⁇ an extended period such as 12 or 24 hours after ingestion.
  • nicotinic acid' * is meant to encompass nicotinic acid or a compound other than nicotinic acid itself which the body metabolizes into nicotinic acid, thus producing essentially the same effect as nicotinic acid.
  • exemplary compounds that produce an effect similar to that of nicotinic acid include, for example, nicotinyl alcohol tartrate, d-glucitol hexanicotinate, aluminum nicotinate, niceritrol and d, 1 -alpha-tocopheryl nicotinate. Each such compound will be collectively referred to herein as "nicotinic acid.”
  • the invention provides a method for treating and/or preventing hyperlipidemia with reduced flushing side effects.
  • the method comprises the steps of administering to a subject in need thereof a therapeutically effective amount of nicotinic acid and a formulation of the invention in an amount sufficient to reduce flushing.
  • the nicotinic acid and/or formulation of the invention may be administered nocturnally.
  • the method involves the use of formulations of the invention to reduce flushing side effects of raloxifene.
  • Raloxifene acts like estrogen in certain places in the body, but is not a hormone. It helps prevent osteoporosis in women who have reached menopause. Osteoporosis causes bones to gradually grow thin, fragile, and more likely to break.
  • Evista slows down the loss of bone mass that occurs with menopause, lowering the risk of spine fractures due to osteoporosis.
  • a common side effect of raloxifene is hot flashes (sweating and flushing). This can be uncomfortable for women who already have hot flashes due to menopause.
  • the method involves the use of formulations of the invention to reduce flushing side effects of antidepressants or anti-psychotic agent.
  • formulations of the invention can be used in conjunction (administered separately or together) with a serotonin reuptake .
  • a 5HT2 receptor antagonist an anticonvulsant, a norepinephrine reuptake inhibitor, an ⁇ -adrenoreceptor antagonist, an NK-3 antagonist, an NK- I receptor antagonist, a PDE4 inhibitor, an Neuropeptide Y5 Receptor Antagonists, a D4 receptor antagonist, a 5HTl A receptor antagonist, a 5HTl D receptor antagonist, a CRF antagonist, a monoamine oxidase inhibitor, or a sedative-hypnotic drug.
  • formulations of the invention may be used as part of a treatment with a serotonin reuptake inhibitor (SRI) to reduce flushing.
  • SRI serotonin reuptake inhibitor
  • the SRI is a selective serotonin reuptake inhibitor (SSRI), such as a fluoxetinoid (fluoxetine, norfluoxetine) or a nefazodoiloid (nefazodone, hydroxynefazodone, oxonefazodone).
  • SSRI selective serotonin reuptake inhibitor
  • SSRI selective serotonin reuptake inhibitor
  • Other exemplary SSRI's include duloxetine, venlafaxine, milnacipran, citalopram, fluvoxamine, paroxetine and sertraline.
  • the formulation of the invention can also be used as part of a treatment with sedative- hypnotic drug, such as selected from the group consisting of a benzodiazepine (such as alprazolam, chlordiazepoxide, clonazepam, chlorazepate, clobazam, diazepam, halazepam, lorazepam, oxazepam and prazepam), Zolpidem, and barbiturates.
  • a formulation of the invention may be used as part of a treatment with a 5-HT1 A receptor partial agonist, such as selected from the group consisting of buspirone, flesinoxan, gepirone and ipsapirone.
  • Formulations of the invention can also used as part of a treatment with a norepinephrine reuptake inhibitor, such as selected from tertiary amine tricyclics and secondary amine .
  • a norepinephrine reuptake inhibitor such as selected from tertiary amine tricyclics and secondary amine .
  • tricyclics such as selected from tertiary amine tricyclics and secondary amine .
  • exemplary tertiary amine tricyclics include amitriptyline, clomipramine, doxepin, imipramine and trimipramine.
  • Exemplary secondary amine tricyclics include amoxapine, desipramine, maprotiline, nortriptyline and protriptyline.
  • formulations of the invention may be used as part of a treatment with a monoamine oxidase inhibitor, such as selected from the group consisting of isocarboxazid, phenelzine, tranylcypromine, selegiline and moclobemide.
  • a monoamine oxidase inhibitor such as selected from the group consisting of isocarboxazid, phenelzine, tranylcypromine, selegiline and moclobemide.
  • formulations of the invention may be used to reduce flushing side effects of chemotherapeutic agents, such as cyclophosphamide, tamoxifen.
  • formulations of the invention may be used to reduce flushing side effects of calcium channel blockers, such as amlodipine.
  • formulations of the invention may be used to reduce flushing side effects of antibiotics.
  • formulations of the invention can be used in combination with levofloxacin.
  • Levofloxacin is used to treat infections of the sinuses, skin, lungs, ears, airways, bones, and joints caused by susceptible bacteria.
  • Levofloxacin also is frequently used to treat urinary infections, including - those resistant to other antibiotics, as well as prostatitis.
  • Levofloxacin is effective in treating infectious diarrheas caused by E. coli, Campylobacter jejuni, and shigella bacteria.
  • Levofloxacin also can be used to treat various obstetric infections, including mastitis.
  • Ocular Disorders One aspect of the present invention is a method for inhibiting, reducing or otherwise treating vision impairment by administering to a patient a therapeutic dosage of a formulation of the invention.
  • the vision impairment is caused by damage to the optic nerve or central nervous system.
  • optic nerve damage is caused by high intraocular pressure, such as that created by glaucoma.
  • optic nerve damage is caused by swelling of the nerve, which is often associated with an infection or an immune (e.g., autoimmune) response such as in optic neuritis.
  • Glaucoma describes a group of disorders which are associated with a visual field defect, cupping of the optic disc, and optic nerve damage. These are commonly referred to as glaucomatous optic neuropathies. Most glaucomas are usually, but not always, associated with a rise in intraocular pressure.
  • Exemplary forms of glaucoma include Glaucoma and Penetrating Keratoplasty, Acute Angle Closure, Chronic Angle Closure, Chronic Open Angle, Angle Recession, Aphakic and Pseudophakic, Drug-Induced, Hyphema, Intraocular Tumors, Juvenile, Lens-Particle, Low Tension, Malignant, Neovascular, Phacolytic, Phacomorphic, Pigmentary, Plateau Iris, Primary Congenital, Primary Open Angle, Pseudoexfoliation, Secondary Congenital, Adult Suspect, Unilateral, Uveitic, Ocular Hypertension, Ocular Hypotony, Posner-Schlossman Syndrome and Scleral Expansion Procedure in Ocular Hypertension & Primary Open-angle Glaucoma.
  • Intraocular pressure can also be increased by various surgical procedures, such as phacoemulsification (i.e., cataract surgery) and implanation of structures such as an artificial lens.
  • phacoemulsification i.e., cataract surgery
  • implanation of structures such as an artificial lens.
  • spinal surgeries in particular, or any surgery in which the patient is prone for an extended period of time can lead to increased interoccular pressure.
  • Optic neuritis is inflammation of the optic nerve and causes acute loss of vision. It is highly associated with multiple sclerosis (MS) as 15-25% of MS patients initially present with ON, and 50-75% of ON patients are diagnosed with MS. ON is also associated with infection (e.g., viral infection, meningitis, syphilis), inflammation (e.g., from a vaccine), infiltration and ischemia.
  • Another condition leading to optic nerve damage is anterior ischemic optic neuropathy (AION).
  • AION anterior ischemic optic neuropathy
  • Arteritic AION is due to giant cell arteritis (vasculitis) and leads to acute vision loss.
  • Non-arteritic AION encompasses all cases of ischemic optic neuropathy other than those due to giant cell arteritis. The pathophysiology of AION is unclear although it appears to incorporate both inflammatory and ischemic mechanisms.
  • optic nerve damage typically associated with demyleination, inflammation, ischemia, toxins, or trauma to the optic nerve.
  • exemplary conditions where the optic nerve is damaged include Demyelinating Optic Neuropathy (Optic Neuritis, Retrobulbar Optic Neuritis), Optic Nerve Sheath Meningioma, Adult Optic Neuritis, Childhood Optic Neuritis, Anterior Ischemic Optic Neuropathy, Posterior Ischemic Optic Neuropathy, Compressive Optic Neuropathy, Papilledema, Pseudopapilledema and Toxic/Nutritional Optic Neuropathy.
  • Demyelinating Optic Neuropathy Optic Neuritis, Retrobulbar Optic Neuritis
  • Optic Nerve Sheath Meningioma Meningioma
  • Adult Optic Neuritis Childhood Optic Neuritis
  • Anterior Ischemic Optic Neuropathy Posterior Ischemic Optic Neuropathy
  • Compressive Optic Neuropathy Papilledema,
  • Other neurological conditions associated with vision loss include Amblyopia, Bells Palsy, Chronic Progressive External Ophthalmoplegia, Multiple Sclerosis, Pseudotumor Cerebri and Trigeminal Neuralgia.
  • the vision impairment is caused by retinal damage.
  • retinal damage is caused by disturbances in blood flow to the eye (e.g., arteriosclerosis, vasculitis).
  • retinal damage is caused by disrupton of the macula (e.g., exudative or non- exudative macular degeneration).
  • Exemplary retinal diseases include Exudative Age Related Macular Degeneration, Nonexudative Age Related Macular Degeneration, Retinal Electronic Prosthesis and RPE Transplantation Age Related Macular Degeneration, Acute Multifocal Placoid Pigment Epitheliopathy, Acute Retinal Necrosis, Best Disease, Branch Retinal Artery Occlusion, Branch Retinal Vein Occlusion, Cancer Associated and Related Autoimmune Retinopathies, Central Retinal Artery Occlusion, Central Retinal Vein Occlusion, Central Serous Chorioretinopathy, Eales Disease, Epimacular Membrane, Lattice Degeneration, Macroaneurysm, Diabetic Macular Edema, Irvine-Gass Macular Edema, Macular Hole, Subretinal Neovascular Membranes, Diffuse Unilateral Subacute Neuroretinitis, Nonpseudophakic Cystoid Macular Edema, Presumed Ocular Histoplasmosis Syndrome,
  • Retinopathy Proliferative Diabetic Retinopathy, Hemoglobinopathies Retinopathy, Purtscher Retinopathy, Valsalva Retinopathy, Juvenile Retinoschisis, Senile Retinoschisis, Terson Syndrome and White Dot Syndromes.
  • exemplary diseases include ocular bacterial infections (e.g. conjunctivitis, keratitis, tuberculosis, syphilis, gonorrhea), viral infections (e.g. Ocular Herpes Simplex Virus, Varicella Zoster Virus, Cytomegalovirus retinitis, Human Immunodeficiency Virus (HIV)) as well as progressive outer retinal necrosis secondary to HIV or other HIV-associated and other immunodeficiency-associated ocular diseases.
  • ocular diseases include fungal infections (e.g. Candida choroiditis, histoplasmosis), protozoal infections (e.g. toxoplasmosis) and others such as ocular toxocariasis and sarcoidosis.
  • One aspect of the invention is a method for inhibiting, reducing or treating vision impairment in a subject undergoing treatment with a chemotherapeutic drug (e.g., a neurotoxic drug, a drug that raises intraocular pressure such as a steroid), by administering to the subject in need of such treatment a therapeutic dosage of a formulation of the invention.
  • a chemotherapeutic drug e.g., a neurotoxic drug, a drug that raises intraocular pressure such as a steroid
  • Another aspect of the invention is a method for inhibiting, reducing or treating vision impairment in a subject undergoing surgery, including ocular or other surgeries performed in the prone position such as spinal cord surgery, by administering to the subject in need of such treatment a therapeutic dosage of a formulation of the invention.
  • Ocular surgeries include cataract, iridotomy and lens replacements.
  • Another aspect of the invention is the treatment, including inhibition and prophylactic treatment, of age related ocular diseases include cataracts, dry eye, retinal damage and the like, by administering to the subject in need of such treatment a therapeutic dosage of a formulation of the invention.
  • age related ocular diseases include cataracts, dry eye, retinal damage and the like, by administering to the subject in need of such treatment a therapeutic dosage of a formulation of the invention.
  • the formation of cataracts is associated with several biochemical changes in the lens of the eye, such as decreased levels of antioxidants ascorbic acid and glutathione, increased lipid, amino acid and protein- oxidation, increased sodium and calcium, loss of amino acids and decreased lens metabolism.
  • the lens which lacks blood vessels, is suspended in extracellular fluids in the anterior part of the eye.
  • Nutrients such as ascorbic acid, glutathione, vitamin E, selenium, bioflavonoids and carotenoids are required to maintain the transparency of the lens.
  • Low levels of selenium results in an increase of free radical-inducing hydrogen peroxide, which is neutralized by the selenium-dependent antioxidant enzyme glutathione peroxidase.
  • Lens-protective glutathione peroxidase is also dependent on the amino acids methionine, cysteine, glycine and glutamic acid.
  • Cataracts can also develop due to an inability to properly metabolize galactose found in dairy products that contain lactose, a disaccharide composed of the monosaccharide galactose and glucose. Cataracts can be prevented, delayed, slowed and possibly even reversed if detected early and metabolically corrected.
  • Retinal damage is attributed, inter alia, to free radical initiated reactions in glaucoma, diabetic retinopathy and age-related macular degeneration (AMD).
  • the eye is a part of the central nervous system and has limited regenerative capability.
  • the retina is composed of numerous nerve cells which contain the highest concentration of polyunsaturated fatty acids (PFA) and subject to oxidation.
  • PFA polyunsaturated fatty acids
  • Free radicals are generated by UV light entering the eye and mitochondria in the rods and cones, which generate the energy necessary to transform light into visual impulses. Free radicals cause peroxidation of the PFA by hydroxyl or superoxide radicals which in turn propagate additional free radicals.
  • the free radicals cause temporary or permanent damage to retinal tissue.
  • Glaucoma is usually viewed as a disorder that causes an elevated intraocular pressure (lOP) that results in permanent damage to the retinal nerve fibers, but a sixth of all glaucoma cases do not develop an elevated IOP.
  • This disorder is now perceived as one of reduced vascular perfusion and an increase in neurotoxic factors.
  • Recent studies have implicated elevated levels of glutamate, nitric oxide and peroxynitirite in the eye as the causes of the death of retinal ganglion cells.
  • Neuroprotective agents may be the future of glaucoma care. For example, nitric oxide synthase inhibitors block the formation of peroxynitrite from nitric oxide and superoxide.
  • Diabetic retinopathy occurs when the underlying blood vessels develop microvascular abnormalities consisting primarily of microaneurysms and intraretinal hemorrhages. Oxidative metabolites are directly involved with the pathogenesis of diabetic retinopathy and free radicals augment the generation of growth factors that lead to enhanced proliferative activity. Nitric oxide produced by endothelial cells of the vessels may also cause smooth muscle cells to relax and result in vasodilation of segments of the vessel. Ischemia and hypoxia of the retina occur after thickening of the arterial basement membrane, endothelial proliferation and loss of pericytes.
  • the inadequate oxygenation causes capillary obliteration or nonperfusion, arteriolar- venular shunts, sluggish blood flow and an impaired ability of RBCs to release oxygen. Lipid peroxidation of the retinal tissues also occurs as a result of free radical damage.
  • the macula is responsible for our acute central vision and composed of light- sensing cells (cones) while the underlying retinal pigment epithelium (RPE) and choroid nourish and help remove waste materials.
  • the RPE nourishes the cones with the vitamin A substrate for the photosensitive pigments and digests the cones shed outer tips.
  • RPE is exposed to high levels of UV radiation, and secretes factors that inhibit angiogenesis.
  • the choroid contains a dense vascular network that provides nutrients and removes the waste materials.
  • the shed cone tips become indigestible by the RPE, where the cells swell and die after collecting too much undigested material. Collections of undigested waste material, called drusen, form under the RPE.
  • Photoxic damage also causes the accumulation of lipofuscin in RPE cells.
  • the intracellular lipofuscin and accumulation of drusen in Bruch's membrane interferes with the transport of oxygen and nutrients to the retinal tissues, and ultimately leads to RPE and photoreceptor dysfunction.
  • blood vessels grow from the choriocapillaris through defects in Bruch's membrane and may grow under the RPE, detaching it from the choroid, and leaking fluid or bleeding.
  • Macular pigment one of the protective factors that prevent sunlight from damaging the retina, is formed by the accumulation of nutritionally derived carotenoids, such as lutein, the fatty yellow pigment that serves as a delivery vehicle for other important nutrients and zeaxanthin.
  • nutritionally derived carotenoids such as lutein, the fatty yellow pigment that serves as a delivery vehicle for other important nutrients and zeaxanthin.
  • Antioxidants such as vitamins C and E, beta-carotene and lutein, as well as zinc, selenium and copper, are all found in the healthy macula. In addition to providing nourishment, these antioxidants protect against free radical damage that initiates macular degeneration.
  • Another aspect of the invention is the prevention or treatment of damage to the eye caused by stress, chemical insult or radiation, by administering to the subject in need of such treatment a therapeutic dosage of a formulation of the invention.
  • a combination drug regimen may include drugs or compounds for the treatment or prevention of ocular disorders or secondary conditions associated with these conditions.
  • a combination drug regimen may include a formulation of the invention and one or more therapeutic agents for the treatment of an ocular disorder.
  • a formulation of the invention can be combined with an effective amount of one or more of: an agent that reduces intraocular pressure, an agent for treating glaucoma, an agent for treating optic - neuritis, an agent for treating CMV Retinopathy, an agent for treating multiple sclerosis, and/or an antibiotic, etc.
  • a formulation of the invention can be administered in conjunction with a therapy for reducing intraocular pressure.
  • One group of therapies involves blocking aqueous production.
  • topical beta-adrenergic antagonists timolol and betaxolol
  • topical timolol causes IOP to fall in 30 minutes with peak effects in 1 -2 hours.
  • Timoptic 0.5% one drop every 30 minutes for 2 doses.
  • the carbonic anhydrase inhibitor, acetazolamide also decreases aqueous production and should be given in conjunction with topical beta-antagonists.
  • An initial dose of 500 mg is administered followed by 250 mg every 6 hours.
  • alpha 2-agonists e.g., Apraclonidine
  • aqueous production e.g., 1,3-bis(trimethyl)-2-agonists
  • e.g., 1,3-bis(trimethyl)-2-agonists act by decreasing aqueous production.
  • Their effects are additive to topically administered beta-blockers. They have been approved for use in controlling an acute rise in pressure following anterior chamber laser procedures, but has been reported effective in treating acute closed-angle glaucoma. A reasonable regimen is 1 drop every 30 minutes for 2 doses.
  • a second group of therapies for reducing intraocular pressure involve reducing vitreous volume.
  • Hyperosmotic agents can be used to treat an acute attack. These agents draw water out of the globe by making the blood hyperosmolar.
  • Oral glycerol in a dose of 1 mL/kg in a cold 50% solution (mixed with lemon juice to make it more palatable) often is used. Glycerol is converted to glucose in the liver; persons with diabetes may need additional insulin if they become hyperglycemic after receiving glycerol.
  • Oral isosorbide is a metabolically inert alcohol that also can be used as an osmotic agent for patients with acute angle-closure glaucoma. Usual dose is 100 g taken p.o.
  • a third group of therapies involve facilitating aqueous outflow from the eye.
  • Miotic agents pull the iris from the iridocorneal angle and may help to relieve the obstruction of the trabecular meshwork by the peripheral iris.
  • Pilocarpine 2% (blue eyes)-4% (brown eyes) can be administered every 15 minutes for the first 1 -2 hours. More frequent administration or higher doses may precipitate a systemic cholinergic crisis.
  • NSAIDS are sometimes used to reduce inflammation.
  • Exemplary therapeutic agents for reducing intraocular pressure include ALPHAGAN® P (Allergan) (brimonidine tartrate ophthalmic solution), AZOPT ⁇ (Alcon) (brinzolamide ophthalmic suspension), BETAGAN® (Allergan) (levobunolol hydrochloride ophthalmic solution, USP), BETIMOL ⁇ (Vistakon) (timolol ophthalmic solution), BETOPTlC S® (Alcon) (betaxolol HCl), BRIMONIDINE TARTRATE (Bausch & Lomb), CARTEOLOL HYDROCHLORIDE (Bausch & Lomb), COSOPT® (Merck) (dorzolamide hydrochloride-timolol maleate ophthalmic solution), LUMIGAN® (Allergan) (bimatoprost ophthalmic solution), OPTIPRANOLOL® (Bausch & Lomb) (metipranolol ophthalmic solution
  • a formulation of the invention can be administered in conjunction with a therapy for treating and/or preventing glaucoma.
  • a glaucoma drug is DARANIDE® Tablets (Merck) (Dichlorphenamide).
  • a formulation of the invention can be administered in conjunction with a therapy for treating and/or preventing optic neuritis.
  • drugs for optic neuritis include DECADRON® Phosphate Injection (Merck) (Dexamethasone Sodium Phosphate), DEPO-MEDROL® (Pharmacia &
  • a formulation of the invention can be administered in conjunction with a therapy for treating and/or preventing CMV Retinopathy.
  • CMV retinopathy treatments for CMV retinopathy include CYTOVENE® (ganciclovir capsules) and VALCYTE® (Roche Laboratories) (valganciclovir hydrochloride tablets).
  • a formulation of the invention can be administered in conjunction with a therapy for treating and/or preventing multiple sclerosis.
  • a therapy for treating and/or preventing multiple sclerosis examples include DANTRIUM® (Procter & Gamble
  • Macrolide antibiotics include tacrolimus, cyclosporine, sirolimus, everolimus, ascomycin, erythromycin, azithromycin, clarithromycin, clindamycin, lincomycin, dirithromycin, josamycin, spiramycin, diacetyl-midecamycin, tylosin, roxithromycin, ABT-773, telithromycin, leucomycins, and lincosamide.
  • Mitochondrial-Associated Diseases and Disorders the invention provides methods for treating diseases or disorders that would benefit from increased mitochondrial activity. The methods involve administering to a subject in need thereof a therapeutically effective amount of a formulation of the invention.
  • Increased mitochondrial activity refers to increasing activity of the mitochondria while maintaining the overall numbers of mitochondria (e.g., mitochondrial mass), increasing the numbers of mitochondria thereby increasing mitochondrial activity (e.g., by stimulating mitochondrial biogenesis), or combinations thereof.
  • diseases and disorders that would benefit from increased mitochondrial activity include diseases or disorders associated with mitochondrial dysfunction.
  • methods for treating diseases or disorders that would benefit from increased mitochondrial activity may comprise identifying a subject suffering from a mitochondrial dysfunction. Methods for diagnosing a mitochondrial dysfunction may involve molecular genetic, pathologic and/or biochemical analysis are summarized in Cohen and Gold, Cleveland Clinic Journal- of Medicine, 68: 625-642 (2001 ).
  • One method for diagnosing a mitochondrial dysfunction is the Thor-Byrne-ier scale (see e.g., Cohen and Gold, supra; Collin S. et al., Eur Neurol. 36: 260-267 (1996)).
  • Other methods for determining mitochondrial number and function include, for example, enzymatic assays (e.g., a mitochondrial enzyme or an ATP biosynthesis factor such as an ETC enzyme or a Krebs cycle enzyme), determination or mitochondrial mass, mitochondrial volume, and/or mitochondrial number, quantification of mitochondrial DNA, monitoring intracellular calcium homeostasis and/or cellular responses to perturbations of this homeostasis, evaluation of response to an apoptogenic stimulus, determination of free radical production.
  • enzymatic assays e.g., a mitochondrial enzyme or an ATP biosynthesis factor such as an ETC enzyme or a Krebs cycle enzyme
  • determination or mitochondrial mass, mitochondrial volume, and/or mitochondrial number quantification of mitochondrial DNA
  • Mitochondria are critical for the survival and proper function of almost all types of eukaryotic cells. Mitochondria in virtually any cell type can have congenital or acquired defects that affect their function. Thus, the clinically significant signs and symptoms of mitochondrial defects affecting respiratory chain function are heterogeneous and variable depending on the distribution of defective mitochondria among cells and the severity of their deficits, and upon physiological demands upon the affected cells. Nondividing tissues with high energy requirements, e.g. nervous tissue, skeletal muscle and cardiac muscle are particularly susceptible to mitochondrial respiratory chain dysfunction, but any organ system can be affected.
  • Diseases and disorders associated with mitochondrial dysfunction include diseases and disorders in which deficits in mitochondrial respiratory chain activity contribute to the development of pathophysiology of such diseases or disorders in a mammal. This includes 1) congenital genetic deficiencies in activity of one or more components of the mitochondrial respiratory chain; and 2) acquired deficiencies in the activity of one or more components of the mitochondrial respiratory chain, wherein such deficiencies are caused by a) oxidative damage during aging; b) elevated intracellular calcium; c) exposure of affected cells to nitric oxide; d) hypoxia or ischemia; e) microtubule-associated deficits in axonal transport of mitochondria, or f) expression of mitochondrial uncoupling proteins.
  • Diseases or disorders that would benefit from increased mitochondrial activity generally include for example, diseases in which free radical mediated oxidative injury leads to tissue degeneration, diseases in which cells inappropriately undergo apoptosis, and diseases in which cells fail to undergo apoptosis.
  • Exemplary diseases or disorders that would benefit from increased mitochondrial activity include, for example, AD (Alzheimer's Disease), ADPD (Alzheimer's Disease and Parkinsons's Disease), AMDF (Ataxia, Myoclonus and Deafness), auto-immune disease, cancer, CIPO (Chronic Intestinal Pseudoobstruction with myopathy and Ophthalmoplegia), congenital muscular dystrophy, CPEO (Chronic Progressive External Ophthalmoplegia), DEAF (Maternally inherited DEAFness or aminoglycoside-induced DEAFness), DEMCHO (Dementia and Chorea), diabetes mellitus (Type I or Type II), DIDMOAD (Diabetes Ins
  • Other diseases and disorders that would benefit from increased mitochondrial activity include, for example, Friedreich's ataxia and other ataxias, amyotrophic lateral sclerosis (ALS) and other motor neuron diseases, macular degeneration, epilepsy, Alpers syndrome, Multiple mitochondrial DNA deletion syndrome, MtDNA depletion syndrome, Complex I deficiency, Complex II (SDH) deficiency, Complex III deficiency, Cytochrome c oxidase (COX, Complex IV) deficiency, Complex V deficiency, Adenine Nucleotide Translocator (ANT) deficiency, Pyruvate dehydrogenase (PDH) deficiency, Ethylmalonic aciduria with lactic acidemia, 3-Methyl glutaconic aciduria with lactic acidemia, Refractory epilepsy with declines during infection, Asperger syndrome with declines during infection, Autism with declines during infection, Attention deficit hyperactivity disorder (ADHD), Cerebral palsy with decline
  • the invention provides methods for treating a subject suffering from mitochondrial disorders arising from, but not limited to, posttraumatic head injury and cerebral edema, stroke (invention methods useful for preventing or preventing reperfusion injury), Lewy body dementia, hepatorenal syndrome, acute liver failure, NASH (non-alcoholic steatohepatitis), Anti- metastasis/prodifferentiation therapy of cancer, idiopathic congestive heart failure, atrial fibrilation (non-valvular), Wolff-Parkinson- White Syndrome, idiopathic heart block, prevention of reperfusion injury in acute myocardial infarctions, familial migraines, irritable bowel syndrome, secondary prevention of non-Q wave myocardial infarctions, Premenstrual syndrome, Prevention of renal failure in hepatorenal syndrome, anti-phospholipid antibody syndrome, eclampsia/pre- eclampsia, oopause infertility, ischemic heart disease/angina, and
  • Types of pharmaceutical agents that are associated with mitochondrial disorders include reverse transcriptase inhibitors, protease inhibitors, inhibitors of DHOD, and the like.
  • reverse transcriptase inhibitors include, for example, Azidothymidine (AZT), Stavudine (D4T), Zalcitabine (ddC), Didanosine (DDI), Fluoroiodoarauracil (FIAU), Lamivudine (3TC), Abacavir and the like.
  • Examples of protease inhibitors include, for example, Ritonavir, Indinavir, Saquinavir, Nelfinavir and the like.
  • inhibitors of dihydroorotate dehydrogenase include, for example, Leflunomide, Brequinar, and the like.
  • Reverse transcriptase inhibitors not only inhibit reverse transcriptase but also polymerase gamma which is required for mitochondrial function. Inhibition of polymerase gamma activity (e.g., with a reverse transcriptase inhibitor) therefore leads to mitochondrial dysfunction and/or a reduced mitochondrial mass which manifests itself in patients as hyperlactatemia. This type of condition may benefit from an increase in the number of mitochondria and/or an improvement in mitochondrial function, e.g., by administration of a formulation of the invention.
  • mitochondrial diseases include cardiomyopathy, muscle weakness and atrophy, developmental delays (involving motor, language, cognitive or executive function), ataxia, epilepsy, renal tubular acidosis, peripheral neuropathy, optic neuropathy, autonomic neuropathy, neurogenic bowel dysfunction, sensorineural deafness, neurogenic bladder dysfunction, dilating cardiomyopathy, migraine, hepatic failure, lactic acidemia, and diabetes mellitus.
  • the invention provides methods for treating a disease or disorder that would benefit from increased mitochondrial activity that involves administering to a subject in need thereof one or more sirtuin activating compounds in combination with another therapeutic agent such as, for example, an agent useful for treating mitochondrial dysfunction (such as antioxidants, vitamins, or respiratory chain cofactors), an agent useful for reducing a symptom associated with a disease or disorder involving mitochondrial dysfunction (such as, an anti- seizure agent, an agent useful for alleviating neuropathic pain, an agent for treating cardiac dysfunction), a cardiovascular agent (as described further below), a chemotherapeutic agent (as described further below), or an anti-neurodegeneration agent (as described further below).
  • an agent useful for treating mitochondrial dysfunction such as antioxidants, vitamins, or respiratory chain cofactors
  • an agent useful for reducing a symptom associated with a disease or disorder involving mitochondrial dysfunction such as, an anti- seizure agent, an agent useful for alleviating neuropathic pain, an agent for treating cardiac dysfunction
  • a cardiovascular agent as
  • the invention provides methods for treating a disease or disorder that would benefit from increased mitochondrial activity that involves administering to a subject in need thereof one or more sirtuin activating compounds in combination with one or more of the following: coenzyme Qjo, L-carnitine, thiamine, riboflavin, niacinamide, folate, vitamin E, selenium, lipoic acid, or prednisone.
  • sirtuin activating compounds in combination with one or more of the following: coenzyme Qjo, L-carnitine, thiamine, riboflavin, niacinamide, folate, vitamin E, selenium, lipoic acid, or prednisone.
  • Compositions comprising such combinations are also provided herein.
  • the invention provides methods for treating diseases or disorders that would benefit from increased mitochondrial acitivty by administering to a subject a therapeutically effective amount of a formulation of the invention.
  • diseases or disorders include, for example, neuromuscular disorders (e.g., Friedreich's Ataxia, muscular dystrophy, multiple sclerosis, etc.), disorders of neuronal instability (e.g., seizure disorders, migrane, etc.), developmental delay, neurodegenerative disorders (e.g., Alzheimer's Disease, Parkinson's Disease, amyotrophic lateral sclerosis, etc.), ischemia, renal tubular acidosis, age-related neurodegeneration and cognitive decline, chemotherapy fatigue, age-related or chemotherapy-induced menopause or irregularities of menstrual cycling or ovulation, mitochondrial myopathies, mitochondrial damage (e.g., calcium accumulation, excitotoxicity, nitric oxide exposure, hypoxia, etc.), and mitochondrial deregulation.
  • neuromuscular disorders e.g., Friedreich's Ataxia
  • FA Friedreich's Ataxia
  • the genetic basis for FA involves GAA trinucleotide repeats in an intron region of the gene encoding frataxin. The presence of these repeats results in reduced transcription and expression of the gene. Frataxin is involved in regulation of mitochondrial iron content.
  • sirtuin activating compounds may be used for treating patients with disorders related to deficiencies or defects in frataxin, including Friedreich's Ataxia, myocardial dysfunction, diabetes mellitus and complications of diabetes like peripheral neuropathy.
  • Muscular dystrophy refers to a family of diseases involving deterioration of neuromuscular structure and function, often resulting in atrophy of skeletal muscle and myocardial dysfunction.
  • Duchenne muscular dystrophy mutations or deficits in a specific protein, dystrophin, are implicated in its etiology. Mice with their dystrophin genes inactivated display some characteristics of muscular dystrophy, and have an approximately 50% deficit in mitochondrial respiratory chain activity. A final common pathway for neuromuscular degeneration in most cases is calcium-mediated impairment of mitochondrial function.
  • sirtuin activating compounds may be used for reducing the rate of decline in muscular functional capacities and for improving muscular functional status in patients with muscular dystrophy.
  • MS Multiple sclerosis
  • MM Multiple sclerosis
  • MM Multiple sclerosis
  • Periodic exacerbations or attacks are significantly correlated with upper respiratory tract and other infections, both bacterial and viral, indicating that mitochondrial dysfunction plays a role in MS.
  • Depression of neuronal mitochondrial respiratory chain activity caused by Nitric Oxide (produced by astrocytes and other cells involved in inflammation) is implicated as a molecular mechanism contributing to MS.
  • sirtuin activating compounds may be used for treatment of patients with multiple sclerosis, both prophylactically and during episodes of disease exacerbation.
  • Epilepsy is often present in patients with mitochondrial cytopathies, involving a range of seizure severity and frequency, e.g.
  • sirtuin activating compounds may be used for treating patients with seizures secondary to mitochondrial dysfunction, including reducing frequency and severity of seizure activity.
  • mitochondrial dysfunction due to either mitochondrial DNA defects or any of the acquired or conditional deficits like exicitoxic or nitric oxide-mediated mitochondrial dysfunction
  • cell proliferation and axonal extension is impaired at crucial stages in development of neuronal interconnections and circuits, resulting in delayed or arrested development of neuropsychological functions like language, motor, social, executive function, and cognitive skills.
  • autism magnetic resonance spectroscopy measurements of cerebral phosphate compounds indicates that there is global undersynthesis of membranes and membrane. precursors indicated by reduced levels of uridine diphospho-sugars, and cytidine nucleotide derivatives involved in membrane synthesis.
  • Disorders characterized by developmental delay include Rett's Syndrome, pervasive developmental delay (or PDD-NOS "pervasive developmental delay not otherwise specified" to distinguish it from specific subcategories like autism), autism,
  • sirtuin activating compounds may be useful for treating treating patients with neurodevelopmental delays (e.g., involving motor, language, executive function, and cognitive skills), or other delays or arrests of neurological and neuropsychological development in the nervous system and somatic development in non-neural tissues like muscle and endocrine glands.
  • neurodevelopmental delays e.g., involving motor, language, executive function, and cognitive skills
  • AD Alzheimer's Disease
  • PD Parkinson's Disease
  • Parkinson's disease but also in peripheral tissues and cells like muscle and platelets of Parkinson's Disease patients.
  • mitochondrial respiratory chain activity is often depressed, especially Complex IV (Cytochrome c Oxidase).
  • mitochondrial respiratory function altogether is depressed as a consequence of aging, further amplifying the deleterious sequelae of additional molecular lesions affecting respiratory chain function.
  • Other factors in addition to primary mitochondrial dysfunction underlie neurodegeneration in AD, PD, and related disorders.
  • Excitotoxic stimulation and nitric oxide are implicated in both diseases, factors which both exacerbate mitochondrial respiratory chain deficits and whose deleterious actions are exaggerated on a background of respiratory chain dysfunction.
  • Huntington's Disease also involves mitochondrial dysfunction in affected brain regions, with cooperative interactions of excitotoxic stimulation and mitochondrial dysfunction contributing to neuronal degeneration.
  • sirtuin activating compounds may be useful for treating and attenuating progression of age-related neurodegenerative diseases including AD and PD.
  • ALS Amyotrophic Lateral Sclerosis
  • SOD 1 Copper-Zinc Superoxide Dismutase
  • Mitochondria both produce and are primary targets for reactive oxygen species. Inefficient transfer of electrons to oxygen in mitochondria is the most significant physiological source of free radicals in mammalian systems. Deficiencies in antioxidants or antioxidant enzymes can result in or exacerbate mitochondrial degeneration. Mice transgenic for mutated SODl develop symptoms and pathology similar to those in human ALS. The development of the disease in these animals has been shown to involve oxidative destruction of mitochondria followed by functional decline of motor neurons and onset of clinical symptoms. Skeletal muscle from ALS patients has low mitochondrial Complex I activity. In certain embodiments, sirtuin activating compounds may be useful for treating ALS, for reversing or slowing the progression of clinical symptoms.
  • Oxygen deficiency results in both direct inhibition of mitochondrial respiratory chain activity by depriving cells of a terminal electron acceptor for
  • Cytochrome c reoxidation at Complex IV and indirectly, especially in the nervous system, via secondary post-anoxic excitotoxicity and nitric oxide formation.
  • tissues are relatively hypoxic.
  • compounds that increase mitochondrial activity provide protection of affected tissues from deleterious effects of hypoxia, attenuate secondary delayed cell death, and accelerate recovery from hypoxic tissue stress and injury.
  • sirtuin activating compounds may be useful for preventing delayed cell death (apoptosis in regions like the hippocampus or cortex occurring about 2 to 5 days after an episode of cerebral ischemia) after ischemic or hypoxic insult to the brain.
  • Acidosis due to renal dysfunction is often observed in patients with mitochondrial disease, whether the underlying respiratory chain dysfunction is congenital or induced by ischemia or cytotoxic agents like cisplatin.
  • Renal tubular acidosis often requires administration of exogenous sodium bicarbonate to maintain blood and tissue pH.
  • sirtuin activating compounds may be useful for treating renal tubular acidosis and other forms of renal dysfunction caused by mitochondrial respiratory chain deficits.
  • mitochondrial respiratory chain function During normal aging, there is a progressive decline in mitochondrial respiratory chain function. Beginning about age 40, there is an exponential rise in accumulation of mitochondrial DNA defects in humans, and a concurrent decline in nuclear-regulated elements of mitochondrial respiratory activity. Many mitochondrial DNA lesions have a selection advantage during mitochondrial turnover, especially in postmitotic cells.
  • mitochondria with a defective respiratory chain produce less oxidative damage to themselves than do mitochondria with intact functional respiratory chains (mitochondrial respiration is the primary source of free radicals in the body). Therefore, normally-functioning mitochondria accumulate oxidative damage to membrane lipids more rapidly than do defective mitochondria, and are therefore "tagged" for degradation by lysosomes.
  • Mitochondrial DNA damage is more extensive and persists longer than nuclear DNA damage in cells subjected to oxidative stress or cancer chemotherapy agents like cisplatin due to both greater vulnerability and less efficient repair of mitochondrial DNA.
  • mitochondrial DNA may be more sensitive to damage than nuclear DNA, it is relatively resistant, in some situations, to mutagenesis by chemical carcinogens. This is because mitochondria respond to some types of mitochondrial DNA damage by destroying their defective genomes rather than attempting to repair them. This results in global mitochondrial dysfunction for a period after cytotoxic chemotherapy.
  • formulations of the invention may be useful for treatment and prevention of side effects of cancer chemotherapy related to mitochondrial dysfunction.
  • a crucial function of the ovary is to maintain integrity of the mitochondrial genome in oocytes, since mitochondria passed onto a fetus are all derived from those present in oocytes at the time of conception. Deletions in mitochondrial DNA become detectable around the age of menopause, and are also associated with abnormal menstrual cycles. Since cells cannot directly detect and respond to defects in mitochondrial DNA, but can only detect secondary effects that affect the cytoplasm, like impaired respiration, redox status, or deficits in pyrimidine synthesis, such products of mitochondrial function participate as a signal for oocyte selection and follicular atresia, ultimately triggering menopause when maintenance of mitochondrial genomic fidelity and functional activity can no longer be guaranteed.
  • Inhibitors of mitochondrial respiration or protein synthesis inhibit hormone-induced ovulation, and furthermore inhibit production of ovarian steroid hormones in response to pituitary gonadotropins.
  • Women with Down's syndrome typically undergo menopause prematurely, and also are subject to early onset of Alzheimer- like dementia.
  • Low activity of cytochrome oxidase is consistently found in tissues of Down's patients and in late-onset Alzheimer's Disease.
  • Appropriate support of mitochondrial function or compensation for mitochondrial dysfunction therefore is useful for protecting against age-related or chemotherapy-induced menopause or irregularities of menstrual cycling or ovulation.
  • formulations of the invention may be useful for treating and preventing amenorrhea, irregular ovulation, menopause, or secondary consequences of menopause.
  • formulations of the invention may be useful for treatment mitochondrial myopathies.
  • Mitochondrial myopathies range from mild, slowly progressive weakness of the extraocular muscles to severe, fatal infantile myopathies and multisystem encephalomyopathies. Some syndromes have been defined, with some overlap between them.
  • Established syndromes affecting muscle include progressive external ophthalmoplegia, the Kearns-Sayre syndrome (with ophthalmoplegia, pigmentary retinopathy, cardiac conduction defects, cerebellar ataxia, and sensorineural deafness), the MELAS syndrome (mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes), the MERFF syndrome (myoclonic epilepsy and ragged red fibers), limb-girdle distribution weakness, and infantile myopathy (benign or severe and fatal). Muscle biopsy specimens stained with modified Gomori's trichrome stain show ragged red fibers due to excessive accumulation of mitochondria.
  • formulations of the invention may be useful for treating patients suffering from toxic damage to mitochondria, such as, toxic damage due to calcium accumulation, excitotoxicity, nitric oxide exposure, drug induced toxic damage, or hypoxia.
  • toxic damage to mitochondria such as, toxic damage due to calcium accumulation, excitotoxicity, nitric oxide exposure, drug induced toxic damage, or hypoxia.
  • Mitochondria are major sites of calcium sequestration, and preferentially utilize energy from the respiratory chain for taking up calcium rather than for ATP synthesis, which results in a downward spiral of mitochondrial failure, since calcium uptake into mitochondria results in diminished capabilities for energy transduction.
  • Excessive stimulation of neurons with excitatory amino acids is a common mechanism of cell death or injury in the central nervous system.
  • Activation of glutamate receptors especially of the subtype designated NMDA receptors, results in mitochondrial dysfunction, in part through elevation of intracellular calcium during excitotoxic stimulation.
  • deficits in mitochondrial respiration and oxidative phosphorylation sensitizes cells to excitotoxic stimuli, resulting in cell death or injury during exposure to levels of excitotoxic neurotransmitters or toxins that would be innocuous to normal cells.
  • Nitric oxide (about 1 micromolar) inhibits cytochrome oxidase (Complex IV) and thereby inhibits mitochondrial respiration; moreover, prolonged exposure to nitric oxide (NO) irreversibly reduces Complex 1 activity. Physiological or pathophysiological concentrations of NO thereby inhibit pyrimidine biosynthesis. Nitric oxide is implicated in a variety of neurodegenerative disorders including inflammatory and autoimmune diseases of the central nervous system, and is involved in mediation of excitotoxic and post-hypoxic damage to neurons.
  • Oxygen is the terminal electron acceptor in the respiratory chain. Oxygen deficiency impairs electron transport chain activity, resulting in diminished pyrimidine synthesis as well as diminished ATP synthesis via oxidative phosphorylation. Human cells proliferate and retain viability under virtually anaerobic conditions if provided with uridine and pyruvate (or a similarly effective agent for oxidizing NADH to optimize glycolytic ATP production).
  • formulations of the invention may be useful for treating diseases or disorders associated with mitochondrial deregulation.
  • mitochondrial DNA encoding respiratory chain components requires nuclear factors. In neuronal axons, mitochondria must shuttle back and forth to the nucleus in order to maintain respiratory chain activity. If axonal transport is impaired by hypoxia or by drugs like taxol which affect microtubule stability, mitochondria distant from the nucleus undergo loss of cytochrome oxidase activity. Accordingly, treatment with a formulation of the invention may be useful for promoting nuclear-mitochondrial interactions.
  • Mitochondria are the primary source of free radicals and reactive oxygen species, due to spillover from the mitochondrial respiratory chain, especially when defects in one or more respiratory chain components impairs orderly transfer of electrons from metabolic intermediates to molecular oxygen.
  • cells can compensate by expressing mitochondrial uncoupling proteins (UCP), of which several have been identified.
  • UCP-2 is transcribed in response to oxidative damage, inflammatory cytokines, or excess lipid loads, e.g. fatty liver and steatohepatitis.
  • UCPs reduce spillover of reactive oxygen species from mitochondria by discharging proton gradients across the mitochondrial inner membrane, in effect wasting energy produced by metabolism and rendering cells vulnerable to energy stress as a trade-off for reduced oxidative injury.
  • Muscle Performance Muscle Performance
  • the invention provides methods for enhancing muscle performance by administering a therapeutically effective amount of a formulation of the invention.
  • formulations of the invention may be useful for improving physical endurance (e.g., ability to perform a physical task such as exercise, physical labor, sports activities, etc.), inhibiting or retarding physical fatigues, enhancing blood oxygen levels, enhancing energy in healthy individuals, enhance working capacity and endurance, reducing muscle fatigue, reducing stress, enhancing cardiac and cardiovascular function, improving sexual ability, increasing muscle ATP levels, and/or reducing lactic acid in blood.
  • the methods involve administering an amount of a formulation of the invention that increase mitochondrial activity, increase mitochondrial biogenesis, and/or increase mitochondrial mass.
  • Sports performance refers to the ability of the athlete's muscles to perform when participating in sports activities. Enhanced sports performance, strength, speed and endurance are measured by an increase in muscular contraction strength, increase in amplitude of muscle contraction, shortening of muscle reaction time between stimulation and contraction. Athlete refers to an individual who participates in sports at any level and who seeks to achieve an improved level of strength, speed and endurance in their performance, such as, for example, body builders, bicyclists, long distance runners, short distance runners, etc. An athlete may be hard training, that is, performs sports activities intensely more than three days a week or for competition. An athlete may also be a fitness enthusiast who seeks to improve general health and well-being, improve energy levels, who works out for about 1-2 hours about 3 times a week. Enhanced sports performance in manifested by the ability to overcome muscle fatigue, ability to maintain activity for longer periods of time, and have a more effective workout.
  • Amino acids including branched-chain amino acids, are released from muscles followed by their deamination to elevate serum ammonia and local oxidation as muscle fuel sources, which augments metabolic acidosis.
  • protein catabolism is initiated where rate of protein synthesis is decreased coupled with an increase in the degradation of non-contractible protein.
  • DGC dystrophin-glycoprotein complex
  • NOS nitric oxide synthase
  • Recent studies of dystrophies related to DGC defects suggest that one mechanism of cellular injury is functional ischemia related to alterations in cellular NOS and disruption of a normal protective action of NO. This protective action is the prevention of local ischemia during contraction-induced increases in sympathetic vasoconstriction. Rando (Microsc Res Tech 55(4):223-35, 2001 ), has shown that oxidative injury precedes pathologic changes and that muscle cells with defects in the DGC have an increased susceptibility to oxidant challenges.
  • the invention provides novel dietary compositions comprising formulations of the invention, a method for their preparation, and a method of using the compositions for improvement of sports performance. Accordingly, provided are therapeutic compositions, foods and beverages that have actions of improving physical endurance and/or inhibiting physical fatigues for those people involved in broadly-defined exercises including sports requiring endurance and labors requiring repeated muscle exertions. Such dietary compositions may additional comprise electrolytes, caffeine, vitamins, carbohydrates, etc. Other Uses
  • Formulations of the invention may be used for treating orpreventing viral infections (such as infections by influenza, herpes or papilloma virus) or as antifungal agents.
  • formulations of the invention may be administered as part of a combination drug therapy with another therapeutic agent for the treatment of viral diseases, including, for example, acyclovir, ganciclovir and zidovudine.
  • formulations of the invention may be administered as part of a combination drug therapy with another anti-fungal agent including, for example, topical anti-fungals such as ciclopirox, clotrimazole, econazole, miconazole, nystatin, oxiconazole, terconazole, and tolnaftate, or systemic anti-fungal such as fluconazole (Diflucan), itraconazole (Sporanox), ketoconazole (Nizoral), and miconazole (Monistat I.V.).
  • topical anti-fungals such as ciclopirox, clotrimazole, econazole, miconazole, nystatin, oxiconazole, terconazole, and tolnaftate
  • systemic anti-fungal such as fluconazole (Diflucan), itraconazole (Sporanox), ketoconazole (Nizoral), and miconazole (Monistat I.V.).
  • Subjects that may be treated as described herein include eukaryotes, such as mammals, e.g., humans, ovines, bovines, equines, porcines, canines, felines, non- human primate, mice, and rats.
  • Cells that may be treated include eukaryotic cells, " e.g., from a subject described above, or plant cells, yeast cells and prokaryotic cells, e.g., bacterial cells.
  • formulations of the invention may be administered to farm animals to improve their ability to withstand farming conditions longer. Formulations of the invention may also be used to increase lifespan, stress resistance, and resistance to apoptosis in plants.
  • a formulation of the invention is applied to plants, e.g., on a periodic basis, or to fungi.
  • plants are genetically modified to produce a compound.
  • plants and fruits are treated with a formulation of the invention prior to picking and shipping to increase resistance to damage during shipping.
  • Plant seeds may also be contacted with a formulation of the invention to preserve them.
  • formulations of the invention may be used for modulating lifespan in yeast cells.
  • Situations in which it may be desirable to extend the lifespan of yeast cells include any process in which yeast is used, e.g., the making of beer, yogurt, and bakery items, e.g., bread.
  • Use of yeast having an extended lifespan can result in using less yeast or in having the yeast be active for longer periods of time.
  • Yeast or other mammalian cells used for recombinantly producing proteins may also be treated as described herein.
  • Formulations of the invention may also be used to increase lifespan, stress resistance and resistance to apoptosis in insects.
  • formulations of the invention would be applied to useful insects, e.g., bees and other insects that are involved in pollination of plants.
  • a formulation of the invention would be applied to bees involved in the production of honey.
  • the methods described herein may be applied to any organism, e.g., eukaryote, that may have commercial importance. For example, they can be applied to fish (aquaculture) and birds (e.g., chicken and fowl).
  • Higher doses of-formulations of the invention may also be used as a pesticide by interfering with the regulation of silenced genes and the regulation of apoptosis during development.
  • a compound may be applied to plants using a method known in the art that ensures the compound is bio-available to insect larvae, and not to plants.
  • formulations of the invention can be applied to affect the reproduction of organisms such as insects, animals and microorganisms.
  • compositions described herein may be formulated in a conventional manner using one or more physiologically acceptable carriers or excipients.
  • compositions may be formulated for administration by, for example, injection (e.g. SubQ, IM, IP), inhalation or insufflation (either through the mouth or the nose) or oral, buccal, sublingual, transdermal, nasal, parenteral or rectal administration.
  • a formulation of the invention may be administered locally, at the site where the target cells are present, i.e., in a specific tissue, organ, or fluid (e.g., blood, cerebrospinal fluid, etc.).
  • the pharmaceutical compositions of the invention are administered orally as a liquid. In other embodiments, the pharmaceutical compositions of the invention are filled into capsules, such as soft gelatin capsules.
  • compositions can be formulated for a variety of modes of administration, including systemic and topical or localized administration. Techniques and formulations generally may be found in Remington's ' Pharmaceutical Sciences, Meade Publishing Co., Easton, PA.
  • parenteral administration injection is preferred, including intramuscular, intravenous, intraperitoneal, and subcutaneous, preferably in physiologically compatible buffers such as Hank's solution or Ringer's solution.
  • Liquid preparations for oral administration may take the form of. for example, solutions, syrups or suspensions. Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives or hydrogenated edible fats); emulsifying agents (e.g., lecithin or acacia); non-aqueous vehicles (e.g., ationd oil, oily esters, ethyl alcohol or fractionated vegetable oils); and preservatives (e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid).
  • the preparations may also contain buffer salts, flavoring, coloring and sweetening agents as appropriate.
  • Preparations for oral administration may be suitably formulated to give controlled release of the active compound.
  • formulations of the invention may be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • a suitable vehicle e.g., sterile pyrogen-free water
  • formulations of the invention may be a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds described herein can be formulated for delivery to the central nervous system (CNS) (reviewed in Begley, Pharmacology & Therapeutics 104: 29-45 (2004)).
  • CNS central nervous system
  • Conventional approaches for drug delivery to the CNS include: neurosurgical strategies (e.g., intracerebral injection or intracerebroventricular infusion); molecular manipulation of the agent (e.g., production of a chimeric fusion protein that comprises a transport peptide that has an affinity for an endothelial cell surface molecule in combination with an agent that is itself incapable of crossing the BBB) in an attempt to exploit one of the endogenous transport pathways of the BBB; pharmacological strategies designed to increase the lipid solubility of an agent (e.g., conjugation of water-soluble agents to lipid or cholesterol carriers); and the transitory disruption of the integrity of the BBB by hyperosmotic disruption (resulting from the infusion of a mannitol solution into the carotid artery or the use of a biologically
  • a pharmaceutical composition described herein is a topical formulation containing a topical carrier that is generally suited to topical drug administration and comprising any such material known in the art.
  • the topical carrier may be selected so as to provide the composition in the desired form, e.g., as an ointment, lotion, cream, microemulsion, gel, oil, solution, or the like, and may be comprised of a material of either naturally occurring or synthetic origin. It is preferable that the selected carrier not adversely affect the active agent or other components of the topical formulation.
  • topical carriers examples include water, alcohols and other nontoxic organic solvents, glycerin, mineral oil, silicone, petroleum jelly, lanolin, fatty acids, vegetable oils, parabens, waxes, and the like. Formulations may be colorless, odorless ointments, lotions, creams, microemulsions and gels.
  • compositions of the invention may be ointments, which generally are semisolid preparations which are typically based on petrolatum or other petroleum derivatives.
  • the specific ointment base to be used is one that will provide for optimum drug delivery, and, preferably, will provide for other desired characteristics as well, e.g., emolliency or the like.
  • an ointment base should be inert, stable, nonirritating and nonsensitizing.
  • ointment bases may be grouped in four classes: oleaginous bases; emulsifiable bases; emulsion bases; and water-soluble bases.
  • Oleaginous ointment bases include, for example, vegetable oils, fats obtained from animals, and semisolid hydrocarbons obtained from petroleum.
  • Emulsifiable ointment bases also known as absorbent ointment bases, contain little or no water and include, for example, hydroxystearin sulfate, anhydrous lanolin and hydrophilic petrolatum.
  • Emulsion ointment bases are either water-in-oil (W/O) emulsions or oil-in-water (O/W) emulsions, and include, for example, cetyl alcohol, glyceryl monostearate, lanolin and stearic acid.
  • Exemplary water-soluble ointment bases are prepared from polyethylene glycols (PEGs) of varying molecular weight; again, reference may be had to Remington's, supra, for further information.
  • compositions of the invention may be creams, which generally are viscous liquid or semisolid emulsions, either oil-in-water or water-in- oil.
  • Cream bases are water-washable, and contain an oil phase, an emulsifier and an aqueous phase.
  • the oil phase is generally comprised of petrolatum and a fatty alcohol such as cetyl or stearyl alcohol; the aqueous phase usually, although not necessarily, exceeds the oil phase in volume, and generally contains a humectant.
  • the emulsifier in a cream formulation as explained in Remington 's, supra, is generally a nonionic, anionic, cationic or amphoteric surfactant.
  • compositions of the invention may be microemulsions, which generally are thermodynamically stable, isotropically clear dispersions of two immiscible liquids, such as oil and water, stabilized by an interfacial film of surfactant molecules (Encyclopedia of Pharmaceutical Technology (New York: Marcel Dekker, 1992), volume 9).
  • surfactant emulsifier
  • co-surfactant co-emulsifier
  • an oil phase and a water phase are necessary.
  • Suitable surfactants include any surfactants that are useful in the preparation of emulsions, e.g., emulsifiers that are typically used in the preparation of creams.
  • the co-surfactant is generally selected from the group of polyglycerol derivatives, glycerol derivatives and fatty alcohols.
  • Preferred emulsifier/co-emulsifier combinations are generally although not necessarily selected from the group consisting of: glyceryl monostearate and polyoxyethylene stearate; polyethylene glycol and ethylene glycol palmitostearate; and caprilic and capric triglycerides and oleoyl macrogolglycerides.
  • the water phase includes not only water but also, typically, buffers, glucose, propylene glycol, polyethylene glycols, preferably lower molecular weight polyethylene glycols (e.g., PEG 300 and PEG 400), and/or glycerol, and the like, while the oil phase will generally comprise, for example, fatty acid esters, modified vegetable oils, silicone oils, mixtures of mono- di- and triglycerides, mono- and di-esters of PEG (e.g., oleoyl macrogol glycerides), etc.
  • buffers glucose, propylene glycol, polyethylene glycols, preferably lower molecular weight polyethylene glycols (e.g., PEG 300 and PEG 400), and/or glycerol, and the like
  • the oil phase will generally comprise, for example, fatty acid esters, modified vegetable oils, silicone oils, mixtures of mono- di- and triglycerides, mono- and di-esters of PEG (e.g., ole
  • compositions of the invention may be gel formulations, which generally are semisolid systems consisting of large organic molecules distributed substantially uniformly throughout a carrier liquid (single phase gels).
  • Single phase gels can be made, for example, by combining the active agent, a carrier liquid and a suitable gelling agent such as tragacanth (at 2 to 5%), sodium alginate (at 2-10%), gelatin (at 2-15%), methylcellulose (at 3-5%), sodium carboxymethylcellulose (at 2-5%), carbomer (at 0.3-5%) or polyvinyl alcohol (at 10-20%) together and mixing until a characteristic semisolid product is produced.
  • suitable gelling agents include methylhydroxycellulose, polyoxyethylene- polyoxypropylene, hydroxyethylcellulose and gelatin.
  • additives may be included in formulations, e.g., topical formulations.
  • additives include, but are not limited to, solubilizers, skin permeation enhancers, opacifiers, preservatives (e.g., anti-oxidants), gelling agents, buffering agents, surfactants (particularly nonionic and amphoteric surfactants), emulsifiers, emollients, thickening agents, stabilizers, humectants, colorants, fragrance, and the like.
  • solubilizers and/or skin permeation enhancers is particularly preferred, along with emulsifiers, emollients and preservatives.
  • An optimum topical formulation comprises approximately: 2 wt. % to 60 wt. %, preferably 2 wt. % to 50 wt. %, solubilizer and/or skin permeation enhancer; 2 wt. % to 50 wt. %, preferably 2 wt. % to 20 wt. %, emulsifiers; 2 wt. %- to 20 wt. % emollient; and 0.01 to 0.2 wt. % preservative, with the active agent and earner (e.g.. water) making of the remainder of the formulation.
  • the active agent and earner e.g.. water
  • a skin permeation enhancer serves to facilitate passage of therapeutic levels of active agent to pass through a reasonably sized area of unbroken skin.
  • Suitable enhancers include, for example: lower alkanols such as methanol ethanol and 2-propanol; alkyl methyl sulfoxides such as dimethylsulfoxide (DMSO), decylmethylsulfoxide (Cm MSO) and tetradecylmethyl sulfboxide; pyrrolidones such as 2-pyrrolidone, N-methyl-2-pyrrolidone and N-(- hydroxyethyl)py ⁇ olidone; urea; N,N-diethyl-m-toluamide; C?
  • miscellaneous solvents such as dimethyl formamide (DMF), N.N- dimethylacetamide (DMA) and tetrahydrofurfuryl alcohol; and the 1 -substituted azacycloheptan-2-ones, particularly 1 -n-dodecylcyclazacycloheptan-2-one (laurocapram; available under the trademark Azone R TM from Whitby Research Incorporated, Richmond, Va.).
  • solubilizers include, but are not limited to, the following: hydrophilic ethers such as diethylene glycol monoethyl ether (ethoxydiglycol, available commercially as Transcutol RTM ) and diethylene glycol monoethyl ether oleate (available commercially as Softcutol RTM ); polyethylene castor oil derivatives such as polyoxy 35 castor oil, polyoxy 40 hydrogenated castor oil, etc.; polyethylene glycol, particularly lower molecular weight polyethylene glycols such as PEG 300 and PEG 400, and polyethylene glycol derivatives such as PEG-8 caprylic/capric glycerides (available commercially as Labrasol RTM ); alkyl methyl sulfoxides such as DMSO; pyrrolidones such as 2-pyrrolidone and N-methyl-2- pyrrolidone; and DMA.
  • hydrophilic ethers such as diethylene glycol monoethyl ether (ethoxydiglycol, available commercially as
  • solubilizers can also act as absorption enhancers.
  • a single solubilizer may be incorporated into the formulation, or a mixture of solubilizers may be incorporated therein.
  • pharmaceutical compositions of the invention exclude solubilizers that act as permeation enhancers, particularly ethoxydiglycol.
  • Suitable emulsifiers and co-emulsifiers include, without limitation, those emulsifiers and co-emulsifiers described with respect to microemulsion formulations.
  • Emollients include, for example, propylene glycol, glycerol, isopropyl myristate, polypropylene glycol-2 (PPG-2) myristyl ether propionate, and the like.
  • sunscreen formulations e.g., other antiinflammatory agents, analgesics, antimicrobial agents, antifungal agents, antibiotics, vitamins, antioxidants, and sunblock agents commonly found in sunscreen formulations including, but not limited to, anthranilates, benzophenones (particularly benzophenone-3), camphor derivatives, cinnamates (e.g., octyl methoxycinnamate), dibenzoyl methanes (e.g., butyl methoxydibenzoyl methane), p-aminobenzoic acid (PABA) and derivatives thereof, and salicylates (e.g., octyl salicylate).
  • sunscreen formulations including, but not limited to, anthranilates, benzophenones (particularly benzophenone-3), camphor derivatives, cinnamates (e.g., octyl methoxycinnamate), dibenzoyl methanes (e.g., but
  • the active agent is present in an amount in the range of approximately 0.25 wt. % to 75 wt. % of the formulation, preferably in the range of approximately 0.25 wt. % to 30 wt. % of the formulation, more preferably in the range of approximately 0.5 wt. % to 15 wt. % of the formulation, and most preferably in the range of approximately 1.0 wt. % to 10 wt. % of the formulation.
  • Topical skin treatment compositions can be packaged in a suitable container to suit its viscosity and intended use by the consumer.
  • a lotion or cream can be packaged in a bottle or a roll-ball applicator, or a propellant-driven aerosol device or a container fitted with a pump suitable for finger operation.
  • the composition When the composition is a cream, it can simply be stored in a non-deformable bottle or squeeze container, such as a tube or a lidded jar.
  • the composition may also be included in capsules such as those described in U.S. Pat. No. 5,063,507.
  • closed containers containing a cosmetically acceptable composition as herein defined.
  • a pharmaceutical formulation for oral or parenteral administration, in which case the formulation may comprises a modulating compound-containing microemulsion as described above, but may contain alternative pharmaceutically acceptable carriers, vehicles, additives, etc. particularly suited to oral or parenteral drug administration.
  • a microemulsion may be administered orally or parenterally substantially as described above, without modification.
  • Pharmaceutical compositions described herein may be stored in oxygen free environment according to methods in the art. For example, resveratrol or analog thereof can be prepared in an airtight capsule for oral administration, such as Capsugel from Pfizer, Inc.
  • the data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans.
  • the dosage of such compounds may lie within a range of circulating concentrations that include the EDso with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the ICso (i.e., the concentration of the test compound that achieves a half-maximal inhibition of symptoms) as determined in cell culture.
  • ICso i.e., the concentration of the test compound that achieves a half-maximal inhibition of symptoms
  • levels in plasma may be measured, for example, by high performance liquid chromatography.
  • kits e.g., kits for therapeutic purposes or kits for modulating the lifespan of cells or modulating apoptosis.
  • a kit may comprise one or more pharmaceutical compositions of the invention, e.g., in premeasured doses.
  • a kit may optionally comprise devices for contacting cells with the compounds and instructions for use. Devices include syringes, stents and other devices for introducing pharmaceutical compositions of the invention into a subject (e.g., the blood vessel of a subject) or applying it to the skin of a subject.
  • the invention provides a composition of matter comprising a formulation of this invention and another therapeutic agent (the same ones used in combination therapies and combination compositions) in separate dosage forms, but associated with one another.
  • a composition of matter comprising a formulation of this invention and another therapeutic agent (the same ones used in combination therapies and combination compositions) in separate dosage forms, but associated with one another.
  • the term "associated with one another" as used herein means that the separate dosage forms are packaged together or otherwise attached to one another such that it is readily apparent that the separate dosage forms are intended to be sold and administered as part of the same regimen.
  • the agent and the formulation of the invention are preferably packaged together in a blister pack or other multi-chamber package, or as connected, separately sealed containers (such as foil pouches or the like) that can be separated by the user (e.g., by tearing on score lines between the two containers).
  • the invention provides a kit comprising in separate vessels, a) a formulation of the invention; and b) another another therapeutic agent such as those described elsewhere in the specification.
  • a liquid formulation was prepared by dissolving resveratrol (drug substance,
  • Emulsion Drug Delivery formulations Two formulations were prepared. One formulation comprises a surfactant mixture forming mixed micelles after dilution with purified water.
  • the first formulation (Formulation 13) has a 5% drug load, contains 45 % polyethyoxylated glycol hydroxystearate (SolutolTMHS15), 5 % PEG-ylated Vitamin E (Vit E TPGS) in a solvent mixture of PEG 300 and EtOH 1/1 w/w.
  • the second formulation forms a micro-emulsion after dilution with purified water and has a 5% drug load, contains 23.75 % medium chain triglyceride (Mygliol 812N), 42.75 % polyethoxylated hydrogenated castor oil (CremophorTM RH40), 4.75% Glyceryl MonoOleate (PeceolTM) and 23.75 % anhydrous ethanol as solvent.
  • Mygliol 812N medium chain triglyceride
  • CremophorTM RH40 polyethoxylated hydrogenated castor oil
  • PeceolTM Glyceryl MonoOleate
  • Polymer formulation The Drug Substance (DS) has a high affinity for polymers relative to other excipients.
  • Polyvinylpyrrolidone (PVP) was found to be a suitable polymer to delay the drug precipitation upon dilution of formulations in which the DS was dissolved, with water.
  • Solid dispersions were produced by solubilizing the DS polymer and surfactant (monoacyl phospholipid or Vit E TGPS or LabrasolTM) in a solvent followed by removal of the solvent.
  • surfactant monoacyl phospholipid or Vit E TGPS or LabrasolTM
  • Table 3 Composition of solid dispersions with resveratrol (Drug Substance (DS) and tendency of precipitation upon hydration/dilution of the formulations in purified water at
  • the drug load was increased to 10% using the same 1/1 w/w ratio of Vit E TPGS/DS and replacing propyleneglycol with LabrasolTM. Upon one-fold dilution with water a slight DS precipitation was observed.
  • Formulation 14 comprised Medium Chain Triglycerides and a 10% drug load (see full composition below), but showed a fast precipitation of the DS upon one- fold dilution with purified water.
  • Formulation 1 1 comprised Long Chain Triglyceride (Soya oil), CapryolTM ⁇ 90, TweenTM 80, GelucireTM 44/14 and ethanol, and showed hardly any precipitation upon one-fold dilution with purified water. After a few hours, some oil droplets were visible on the surface of the water phase. To further improve formulation Formulation 1 1 , the use of the two lipids
  • GelucireTM 44/14 and LabrasolTM were explored as main components in the formulations. It was found that when DS was dissolved in GelucireTM 44/14 at 100 mg/g, the drug precipitated in a fast manner after dilution with purified water. At a concentration of 150 mg DS/g In LabrasolTM (Formulation 10), no precipitation of crystalline drug was observed upon dilution with purified water. The DS precipitated in the form of oil-like droplets. With a combination of both excipients, a drug load of 10% DS could be achieved without the need of ethanol. Higher drug loads can be achieved by using the two lipids in combination with ethanol as additional solvent/excipient to fully solubilize the drug in the formulation.
  • the Formulation 10 showed an increase of degradation product of 0.1 % and 0.5% after 1 -week storage at 25°C and 40 0 C, respectively. It is important to note that the color of the formulation changed from a slight yellowish color at TO to a slight pink and dark red at 25°C and 40 0 C, respectively. Formulation 1 1 also showed a color change at 25°C and 40 0 C compared to the initial color but no increase of degradation products was observed.
  • the objective of this study was to assess the pharmacokinetics of resveratrol in 3 formulations in C57BL/6 mice following oral administration.
  • 72 C57BL/6 mice were divided into 3 treatment groups, 24 per group.
  • Test compound resveratrol was dissolved in vehicles and orally administrated to those mice at 500 mg/kg.
  • Serial plasma samples were collected following oral administration, after which resveratrol concentration in plasma were determined and pharmacokinetic property in mice were evaluated.
  • resveratrol 400 mg were weighed into a mortar and pestle for grinding. To this was added 7.6 g of lipid matrix vehicle 1 (60% monoacyl phospholipid, 20% PEG300, 20% propyleneglycol) and the contents were stirred rapidly. After stirring homogenously, 0.9% saline was added to lower viscosity and the container was rinsed with 0.9% saline, and the mixture was vortexed thoroughly to yield a homogenous suspension at 25 mg/mL. Animals were dosed orally at 20 ml/kg to achieve an oral dose of 500 mg/kg.
  • lipid matrix vehicle 1 70% monoacyl phospholipid, 20% PEG300, 20% propyleneglycol
  • resveratrol 400 mg were weighed into a vial for mixing. 7.6 g from the vial labeled emulsion vehicle (45% polyethyoxylated glycol hydroxystearate (SolutolTM HS 15); 5.0% PEG-ylated Vitamin E (Vit E TPGS); 50% PEG300/ EtOH (1/1 w/w) were added and the mixture was vortexed until complete dissolution of the drug substance at 50 mg/mL by ultrasonic. Animals were dosed orally at 10 ml/kg to achieve an oral dose of 500 mg/kg.
  • emulsion vehicle 45% polyethyoxylated glycol hydroxystearate (SolutolTM HS 15); 5.0% PEG-ylated Vitamin E (Vit E TPGS); 50% PEG300/ EtOH (1/1 w/w) were added and the mixture was vortexed until complete dissolution of the drug substance at 50 mg/mL by ultrasonic. Animals were dosed orally at 10 m
  • lipid matrix vehicle 2 25% medium chain triglyceride (Mygliol TM 812N); 45% polyethoxylated hydrogenated castor oil (CremaphorTM RH40); 5.0% Glycerl MonoOleate (PeceolTM); 25% ethanol) were added and the mixture was vortexed until complete dissolution of the drug substance at 50 mg/mL by ultrasonic. Animals were dosed orally at 10 ml/kg to achieve an oral dose of 500 mg/kg.
  • the concentrations of resveratrol in plasma were determined using a high performance liquid chromatography/mass spectrometry (HPLC/MS/MS) method.
  • Plasma samples were processed by liquid-liquid extraction. Plasma samples (0.1 niL) were transferred to Eppendorff tube, then 20 ⁇ L methanol, 50 ⁇ L internal standard solution (1 ⁇ g/mL) and 500 ⁇ L methyl tert-butyl ether (MTBE) were added to it.
  • Internal standard solution was prepared by dissolving IS compound (CAS: 2896-608) in H 2 O to yield a final concentration of 200 ⁇ g/mL, then diluted to 1 ⁇ g/mL. After vortexing for 1 min and centrifuging for 5 min at 15,000 rpm, 450 ⁇ L of supernatant was transferred to a glass tube and evaporated under vacuum condition. The residue was reconstituted with 100 ⁇ L methanol
  • the LC system comprised an Agilent (Agilent Technologies Inc. USA) liquid chromatograph equipped with an isocratic pump ( 1 100 series), an autosampler ( 1 100 series) and a degasser (1 100 series). Mass spectrometric analysis was performed using an AP13000 (triple-quadrupole) instrument from AB Inc (Canada) with an ESI interface. The data acquisition and control system were created using Analyst 1.4 software from ABI Inc.
  • the values of C 1113x and T 1113x for resveratrol were 9145.49 ⁇ g/L, 0.08 h, respectively, while the values of AUQo-t) and Ti/ 2 were 5457.66 h* ⁇ g/L and 4.08 h, respectively.
  • the values of C max and T 1113x for resveratrol were 5431 .25 ⁇ g/L, 0.08 h, respectively, while the values of AUQo-o and T ⁇ a were 8325.98 h* ⁇ g/L and 1 .60 h, respectively.
  • Dawley rats males, 8- 10 weeks old, 200 to 250gr were divided into 4 treatment groups, 18 per group.
  • Test compound resveratrol was dissolved in vehicles and orally administrated to the rats at 100 or 500 mg/kg.
  • Serial plasma samples were collected following oral administration, after which resveratrol concentration in plasma were determined and pharmacokinetic property in mice were evaluated.
  • the blood was collected at 0 (pre-dosing) and 5 min, 15 min, 30 min, 1 hr, 2 hr, 4 hr, 6 hr, 8 hr and 24 hr post administration. Blood samples for each time point were collected within 10% for the time points before the first hour and within ⁇ 2 min for the time points after 1 hour. The plasma was separated via centrifugation within 1 hr and stored in -20°C before analysis. Analysis was done as before for the mouse study in the previous example. Results are shown in Table 7.
  • Formula 15 was prepared as follows. Resveratrol (99% Grade from PCAS, Lima France) was filled into 00 capsules (Torpac USP Grade gelatin capsules) using a Profill Capsule Filling System (Torpac, Model No. FB-2783). 100 capsules were flush filled with resveratrol according to the filling system operating instructions. The resulting filled capsules were visually inspected and weighed. Average net capsule weight was 544.1 mg/capsule (standard deviation was 16.6 mg/capsule). 5 capsules are approximately the equivalent of 2.7 grams of resveratrol.
  • Formula 10 was also prepared as soft gel capsules (Formula 10a).
  • the liquid capsules for dosing were produced by manufacturing a 300 g batch of the resveratrol fill formulation, filling that formulation into liquid capsules and then sealing the capsules. Specifically, 30% resveratrol was added to 70% caprylocaproyl macrogol 8 glycerides (Labrasol, Gattefosse) and mixed at 40 0 C for 60 minutes (all percentages are on a weight basis). The resulting fill formulation is an opaque, homogenous suspension. The fill formulation was continually mixed while cooling to 30 0 C and then transferred to the filling hopper for capsule filling. Specific gravity of the mixture was 1.1 (at 25°C).
  • the fill product hopper temperature was set at 30°C, and the drying air temperature was set to 40 0 C.
  • Liquid gelatin capsules (Size 00 Licaps HGC Natural/Natural, Capsugel) were filled and sealed on a CFS 1200 Capsule Filling and Sealing Machine (Capsugel) according to the equipments standard operating procedures. The capsules were filled volumetrically with 0.75 ⁇ 0.10 mL of the fill formulation. The theoretical concentration of resveratrol in each capsule was 250 mg/capsule. Seven capsules were orally dosed to delivery 1.7 g of active resveratrol. The study was carried out in 4 subjects who received different resveratrol formulations by oral administration.
  • resveratrol 1.7 or 2.7 g
  • Administration of resveratrol took place approximately 15 min after subjects ingested a standard breakfast.
  • Blood samples were collected to assess the PK parameters of each formulation at the following time points: Baseline (pre-administration), 30 and 60 minutes, 2 hr, 4 hr and 24 hr.
  • Blood samples for PK analysis were collected in EDTA vacutainers ( ⁇ 5 mL per draw).
  • Plasma concentrations of resveratrol were estimated using a validated LCMS/MS method. Tmax, Cmax, and AUC(0-4hr) were evaluated using standard statistical software. Results are shown in Table 8 and Figure 1.
  • Formulation 10 provides significant exposure (287% increase in AUC and 186% in Cmax) of Resveratrol compared to the reference Resveratrol capsule.
  • Table 8. Selected Pharmacokinetics Parameters of Resveratrol in Humans Following Oral Administration in Three Formulations at Indicated Dose
  • any polynucleotide and polypeptide sequences which reference an accession number correlating to an entry in a public database, such as those maintained by The Institute for Genomic Research (TIGR) (www.tigr.org) and/or the National Center for Biotechnology Information (NCBl) (www.ncbi.nlm.nih.gov).
  • TIGR The Institute for Genomic Research
  • NCBl National Center for Biotechnology Information

Abstract

Resveratrol can be used to treat or inhibit the onset of many diseases that are related to the aging process, but large doses are required. The present invention provides concentrated liquid formulations of resveratrol, typically having a resveratrol concentration of at least 10% by weight. These formulations improve patient compliance because the patient need not consume such a large volume of a resveratrol formulation.

Description

RESVERATROL FORMULATIONS
RELATED APPLICATION
This application claims the benefit of U. S Provisional Application No. 61/010,505, filed January 8, 2008, the contents of which are incorporated herein by reference in their entirety.
BACKGROUND OF THE INVENTION
The Silent Information Regulator (SIR) family of genes represents a highly conserved group of genes present in the genomes of organisms ranging from archaebacteria to a variety of eukaryotes (Frye, 2000). The encoded SIR proteins are involved in diverse processes from regulation of gene silencing to DNA repair. The proteins encoded by members of the SIR2 gene family show high sequence conservation in a 250 amino acid core domain. A well-characterized gene in this family is S. cerevisiae SIR2, which is involved in silencing HM loci that contain information specifying yeast mating type, telomere position effects and cell aging (Guarente, 1999; Kaeberlein et al., 1999; Shore, 2000). The yeast Sir2 protein belongs to a family of histone deacetylases (reviewed in Guarente, 2000; Shore, 2000). The Sir2 homolog, CobB, in Salmonella typhimurium, functions as an NAD (nicotinamide adenine dinucleotide)-dependent ADP-ribosyl transferase (Tsang and Escalante-Semerena, 1998).
The Sir2 protein is a deacetylase which uses NAD as a cofactor (Imai et al., 2000; Moazed, 2001 ; Smith et al., 2000; Tanner et al., 2000; Tanny and Moazed, 2001 ). Unlike other deacetylases, many of which are involved in gene silencing, Sir2 is insensitive to histone deacetylase inhibitors like trichostatin A (TSA) (Imai et al., 2000; Landry et al., 2000a; Smith et al., 2000).
Deacetylation of acetyl-lysine by Sir2 is tightly coupled to NAD hydrolysis, producing nicotinamide and a novel acetyl-ADP ribose compound (1 -O-acetyl- ADP-ribose) (Tanner et al., 2000; Landry et al., 2000b; Tanny and Moazed, 2001 ). The NAD-dependent deacetylase activity of Sir2 is essential for its functions which can connect its biological role with cellular metabolism in yeast (Guarente, 2000; Imai et al., 2000; Lin et al., 2000; Smith et al., 2000). Mammalian Sir2 homologs have NAD-dependent histone deacetylase activity (Imai et al., 2000; Smith et al., 2000). Most information about Sir2 mediated functions comes from the studies in yeast (Gartenberg, 2000; Gottschling, 2000).
Biochemical studies have shown that Sir2 can readily deacetylate the amino- terminal tails of histones H3 and H4, resulting in the formation of 1 -0-acetyl-ADP- ribose and nicotinamide. Strains with additional copies of SIR2 display increased rDNA silencing and a 30% longer life span. It has recently been shown that additional copies of the C. elegans SIR2 homolog, sir-2.1, greatly extend life span in that organism. This implies that the £//?2-dependent regulatory pathway for aging arose early in evolution and has been well conserved. Yeast life span, like that of metazoans, is also extended by interventions that resemble caloric restriction. Mutations that reduce the activity of the glucose-responsive cAMP (adenosine 3'5'- monophosphate)-dependent (PKA) pathway extend life span in wild type cells but not in mutant sir2 strains, demonstrating that S1R2 is a key downstream component of the caloric restriction pathway.
Recently, a number of small molecule activators of the SIR proteins, such as resveratrol, have been reported and a number of uses for these compounds have been identified. For example, small molecule activators of SIR proteins were shown to extend life span in yeast and cultured human cells as well as activate SIR protein activity in human cells {supra). Additionally, the small molecule SIR activators were shown to mimic calorie restriction and extend lifespan in Caenorhabditis elegans and Drosophila melanogaster {supra). Activators of the SIR proteins may therefore be useful for mimicking the effects of calorie restriction in eukaryotic cells and treating aging-related diseases such as stroke, cardiovascular disease, arthritis, high blood pressure, or Alzheimer's disease {supra). Additionally, it has been shown that resveratrol promotes fat mobilization in C. elegans, prevent fat accumulation in C. elegans, stimulate fat mobilization in mammalian cells, and inhibit adipogenesis in mammalian cells (see e.g., U.S. Patent Publication No. 2005/0171027 and PCT Publication No. WO 2005/065667). Additionally, resveratrol was shown to at least partially restore insulin sensitivity in insulin resistant cells {supra).
In order to attain the benefits seen in cells in a whole organism, it would be advantageous to have a formulation of resveratrol that could deliver a sufficient dose of the agent in a convenient form. In particular, it would be advantageous to have a highly concentrated liquid formulation of resveratrol, where the resveratrol stably remained in solution.
SUMMARY OF THE INVENTION
Applicants have discovered several liquid formulations of resveratrol (e.g., solutions, emulsions), where the resveratrol is in solution and is highly concentrated. Typically, liquid formulations of the invention include at least 1 1% or even 15% by weight of resveratrol. The invention also includes methods of preparing these formulations. In addition, the invention includes methods of treating a variety of conditions using the formulations described herein. Also, the invention includes the use of formulations of the invention for treating the conditions and diseases described herein, along with the use of the formulations in the manufacture of a medicament for treating the conditions and diseases described herein.
BRIEF DESCRIPTION OF THE DRAWING
Figure 1 shows in vivo pharmacokinetic profiles of various resveratrol formulations administered to healthy human volunteers.
DETAILED DESCRIPTION OF THE INVENTION 1. Definitions
As used herein, the following terms and phrases shall have the meanings set forth below. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art. The term "agent" is used herein to denote a chemical compound, a mixture of chemical compounds, a biological macromolecule (such as a nucleic acid, an antibody, a protein or portion thereof, e.g., a peptide), or an extract made from biological materials such as bacteria, plants, fungi, or animal (particularly mammalian) cells or tissues. The activity of such agents may render it suitable as a "therapeutic agent" which is a biologically, physiologically, or pharmacologically active substance (or substances) that acts locally or systemically in a subject. The term "bioavailable" when referring to a compound is art-recognized and refers to a form of a compound that allows for it, or a portion of the amount of compound administered, to be absorbed by, incorporated to, or otherwise physiologically available to a subject or patient to whom it is administered. The term "companion animals" refers to cats and dogs. As used herein, the term "dog(s)" denotes any member of the species Canis familiaris, of which there are a large number of different breeds. The term "cat(s)" refers to a feline animal including domestic cats and other members of the family Felidae, genus Felis.
"Diabetes" refers to high blood sugar or ketoacidosis, as well as chronic, general metabolic abnormalities arising from a prolonged high blood sugar status or a decrease in glucose tolerance. "Diabetes" encompasses both the type I and type II (Non Insulin Dependent Diabetes Mellitus or NIDDM) forms of the disease. The risk factors for diabetes include the following factors: waistline of more than 40 inches for men or 35 inches for women, blood pressure of 130/85 mmHg or higher, triglycerides above 150 mg/dl, fasting blood glucose greater than 100 mg/dl or high- density lipoprotein of less than 40 mg/dl in men or 50 mg/dl in women.
The term "hyperinsulinemia" refers to a state in an individual in which the level of insulin in the blood is higher than normal.
The term "insulin resistance" refers to a state in which a normal amount of insulin produces a subnormal biologic response relative to the biological response in a subject that does not have insulin resistance.
An "insulin resistance disorder," as discussed herein, refers to any disease or condition that is caused by or contributed to by insulin resistance. Examples include: diabetes, obesity, metabolic syndrome, insulin-resistance syndromes, syndrome X, insulin resistance, high blood pressure, hypertension, high blood cholesterol, dyslipidemia, hyperlipidemia, dyslipidemia, atherosclerotic disease including stroke, coronary artery disease or myocardial infarction, hyperglycemia, hyperinsulinemia and/or hyperproinsulinemia, impaired glucose tolerance, delayed insulin release, diabetic complications, including coronary heart disease, angina pectoris, congestive heart failure, stroke, cognitive functions in dementia, retinopathy, peripheral neuropathy, nephropathy, glomerulonephritis, glomerulosclerosis, nephrotic syndrome, hypertensive nephrosclerosis some types of cancer (such as endometrial, breast, prostate, and colon), complications of pregnancy, poor female reproductive health (such as menstrual irregularities, infertility, irregular ovulation, polycystic ovarian syndrome (PCOS)), lipodystrophy, cholesterol related disorders, such as gallstones, cholescystitis and cholelithiasis, gout, obstructive sleep apnea and respiratory problems, osteoarthritis, and prevention and treatment of bone loss, e.g. osteoporosis.
The term "livestock animals" refers to domesticated quadrupeds, which includes those being raised for meat and various byproducts, e.g., a bovine animal including cattle and other members of the genus Bos, a porcine animal including domestic swine and other members of the genus Sus, an ovine animal including sheep and other members of the genus Ovis, domestic goats and other members of the genus Capra; domesticated quadrupeds being raised for specialized tasks such as use as a beast of burden, e.g., an equine animal including domestic horses and other members of the family Equidae, genus Equus. The term "mammal" is known in the art, and exemplary mammals include humans, primates, livestock animals (including bovines, porcines, etc.), companion animals (e.g., canines, felines, etc.) and rodents (e.g., mice and rats).
"Obese" individuals or individuals suffering from obesity are generally individuals having a body mass index (BMI) of at least 25 or greater. Obesity may or may not be associated with insulin resistance.
The terms "parenteral administration" and "administered parenterally" are art-recognized and refer to modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intra- articulare, subcapsular, subarachnoid, intraspinal, and intrasternal injection and infusion.
A "patient", "subject", "individual" or "host" refers to either a human or a non-human animal. The term "pharmaceutically acceptable carrier" is art-recognized and refers to a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting any subject composition or component thereof. Each carrier must be "acceptable" in the sense of being compatible with the subject composition and its components and not injurious to the patient. Some examples of materials which may serve as pharmaceutically acceptable carriers include: (1 ) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (1 1 ) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21 ) other non-toxic compatible substances employed in pharmaceutical formulations.
The term "phospholipids" refers to lipids that contain a phosphate group, typically attached to the glycerol. Examples of phospholipids include lecithin, hydroxylated lecithin, Lysophosphatidylcholine, Cardiolipin, Sphingomyelin, Phosphatidylcholine, Phosphatidyl ethanolamine, Phosphatidic acid, Phosphatidyl glycerol and Phosphatidyl serine.
The term "polar lipid" refers to a lipid having a full or partial positive or negative charge. Such lipids are typically surfactants.
The term "prophylactic" or "therapeutic" treatment is art-recognized and refers to administration of a drug to a host. If it is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the host animal) then the treatment is prophylactic, i.e., it protects the host against developing the unwanted condition, whereas if administered after manifestation of the unwanted condition, the treatment is therapeutic (i.e., it is intended to diminish, ameliorate or maintain the existing unwanted condition or side effects therefrom). "Replicative lifespan" of a cell refers to the number of daughter cells produced by an individual "mother cell." "Chronological aging" or "chronological lifespan," on the other hand, refers to the length of time a population of non- dividing cells remains viable when deprived of nutrients. "Increasing the lifespan of a cell" or "extending the lifespan of a cell," as applied to cells or organisms, refers to increasing the number of daughter cells produced by one cell; increasing the ability of cells or organisms to cope with stresses and combat damage, e.g., to DNA, proteins; and/or increasing the ability of cells or organisms to survive and exist in a living state for longer under a particular condition, e.g., stress (for example, heatshock, osmotic stress, high energy radiation, chemically-induced stress, DNA damage, inadequate salt level, inadequate nitrogen level, or inadequate nutrient level). Lifespan can he increased by at least about 20%, 30%, 40%, 50%, 60% or between 20% and 70%, 30% and 60%, 40% and 60% or more using methods described herein.
"Sirtuin-activating compound" refers to a compound that increases the level of a sirtuin protein and/or increases at least one activity of a sirtuin protein. In an exemplary embodiment, a sirtuin-activating compound may increase at least one biological activity of a sirtuin protein by at least about 10%, 25%, 50%, 75%, 100%, or more. Exemplary biological activities of sirtuin proteins include deacetylation, e.g., of histones and p53; extending lifespan; increasing genomic stability; silencing transcription; and controlling the segregation of oxidized proteins between mother and daughter cells. "Sirtuin protein" refers to a member of the sirtuin deacetylase protein family, or preferably to the sir2 family, which include yeast Sir2 (GenBank Accession No. P53685), C. elegans Sir-2.1 (GenBank Accession No. NP_501912), and human SIRTl (GenBank Accession No. NMJ)12238 and NP_036370 (or AF083106)) and SIRT2 (GenBank Accession No. NMJ312237, NM_030593, NP_036369, NP_085096, and AF083107) proteins. Other family members include the four additional yeast Sir2-like genes termed "HST genes" (homologues of Sir two) HSTl , HST2, HST3 and HST4, and the five other human homologues hSIRT3, hSIRT4, hSIRT5, hSIRT6 and hSIRT7 (Brachmann et al. (1995) Genes Dev. 9:2888 and Frye et al. (1999) BBRC 260:273). Preferred sirtuins are those that share more similarities with SIRTl , i.e., hSIRTl , and/or Sir2 than with SIRT2, such as those members having at least part of the N-terminal sequence present in SIRTl and absent in SIRT2 such as SIRT3 has. "SIRTl protein" refers to a member of the sir2 family of sirtuin deacetylases. In one embodiment, a SIRTl protein includes yeast Sir2 (GenBank Accession No. P53685), C. elegans Sir-2.1 (GenBank Accession No. NP_501912), human SIRTl (GenBank Accession No. NM_O12238 or NP_036370 (or AF083106)), and human SIRT2 (GenBank Accession No. NM Ol 2237, NM_030593, NP 036369,
NP 085096, or AF083107) proteins, and equivalents and fragments thereof. In another embodiment, a SIRTl protein includes a polypeptide comprising a sequence consisting of, or consisting essentially of, the amino acid sequence set forth in GenBank Accession Nos. NP 036370, NP_501912, NP_085096, NP 036369, or P53685. SIRTl proteins include polypeptides comprising all or a portion of the amino acid sequence set forth in GenBank Accession Nos. NP 036370, NP 501912, NPJD85096, NP 036369, or P53685; the amino acid sequence set forth in GenBank Accession Nos. NP_036370, NP_501912, NP_085096, NP_036369, or P53685 with 1 to about 2, 3, 5, 7, 10, 15, 20, 30, 50, 75 or more conservative amino acid substitutions; an amino acid sequence that is at least 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, or 99% identical to GenBank Accession Nos. NP_036370, NP 501912, NP_085096, NP 036369, or P53685, and functional fragments thereof. Polypeptides of the invention also include homologs (e.g., orthologs and paralogs), variants, or fragments, of GenBank Accession Nos. NP 036370, NP_501912, NP_085096, NP_036369, or P53685.
A surfactant is a compound having both polar (hydrophilic) and nonpolar (hydrophobic) regions. The polar region of a surfactant may be ionic (positive or negative) or nonionic. Surfactants are useful in preparing emulsions and in solubilizing substances. The terms "systemic administration," "administered systemically,"
"peripheral administration" and "administered peripherally" are art-recognized and refer to the administration of a subject composition, therapeutic or other material other than directly into the central nervous system, such that it enters the patient's system and, thus, is subject to metabolism and other like processes. The term "therapeutic agent" is art-recognized and refers to any chemical moiety that is a biologically, physiologically, or pharmacologically active substance that acts locally or systemically in a subject. The term also means any substance intended for use in the diagnosis, cure, mitigation, treatment or prevention of disease or in the enhancement of desirable physical or mental development and/or conditions in an animal or human.
The term "therapeutic effect" is art-recognized and refers to a local or systemic effect in animals, particularly mammals, and more particularly humans caused by a pharmacologically active substance. The phrase "therapeutically- effective amount" means that amount of such a substance that produces some desired local or systemic effect at a reasonable benefit/risk ratio applicable to any treatment. The therapeutically effective amount of such substance will vary depending upon the subject and disease condition being treated, the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art. For example, certain compositions described herein may be administered in a sufficient amount to produce a desired effect at a reasonable benefit/risk ratio applicable to such treatment.
"Treating" a condition or disease refers to curing as well as ameliorating at least one symptom of the condition or disease.
The term "vision impairment" refers to diminished vision, which is often only partially reversible or irreversible upon treatment (e.g., surgery). Particularly severe vision impairment is termed "blindness" or "vision loss", which refers to a complete loss of vision, vision worse than 20/200 that cannot be improved with corrective lenses, or a visual field of less than 20 degrees diameter (10 degrees radius).
2. Resveratrol Formulations
In one aspect, the invention provides novel pharmaceutical compositions (also referred to as "formulations") for treating and/or preventing a wide variety of diseases and disorders including, for example, diseases or disorders related to aging or stress, diabetes, obesity, neurodegenerative diseases, ocular diseases and disorders, cardiovascular diseases, blood clotting disorders, inflammation, cancer, and/or flushing, etc. Pharmaceutical compositions of the invention may also be used for treating a disease or disorder in a subject that would benefit from increased mitochondrial activity, for enhancing muscle performance, for increasing muscle ATP levels, or for treating or preventing muscle tissue damage associated with hypoxia or ischemia.
Resveratrol is 3,5,4'-trihydroxystilbene and has both cis and trans isomers. The structure of each isomer is shown below:
.
The invention also includes salts and esters of resveratrol, where indicated. Base addition salts include those derived from inorganic bases, such as ammonium or alkali or alkaline earth metal hydroxides, carbonates, bicarbonates, and the like. Such bases useful in preparing the salts of this invention thus include sodium hydroxide, potassium hydroxide, ammonium hydroxide, potassium carbonate, and the like.
The compounds and salts and esters thereof can be present in amorphous or crystalline (including co-crystalline and polymorph) forms, unless otherwise indicated.
In certain embodiments, the liquid formulations of the invention (e.g., solutions, emulsions) include at least 1 1% by weight of resveratrol or a salt or ester thereof. In certain such embodiments, the formulation includes at least 12%, at least 13%, at least 14%, at least 15%, at least 16%, at least 17%, at least 1 8%, at least 19%, at least 20%, at least 21 %, at least 22%, at least 23%, at least 24% or at least 25% of resveratrol (or its salt or ester).
In certain embodiments, the formulation includes resveratrol and one or more of monoglycerides, diglycerides, triglycerides, fatty acid monoesters of polyethylene glycol (PEG) and fatty acid diesters of PEG. Typically, the monoglycerides, diglycerides, triglycerides, fatty acid monoesters of PEG and fatty acid diesters of PEG include C8 and C l O fatty acids, such as caprylocapryol macrogol glycerides. In a particular embodiment, the formulation is a solution and the solution consists essentially of resveratrol and caprylocapryol macrogol glycerides (commercially available as Labrasol™).
Such formulations may also include a polar lipid, such as a phospholipid (e.g., lecithin). In such embodiments, the amount of polar lipid is typically from 0.25-5% (e.g., by weight), such as 1-3% or 1.5-2.5%, with the remainder resveratrol and monoglycerides, diglycerides, triglycerides, fatty acid monoesters of polyethylene glycol (PEG) and fatty acid diesters of PEG.
Alternatively, such formulations may also include one or more of saturated polyglycolized glycerides (e.g., Gelucire 44/14) and ethanol. In certain embodiments, the amount of saturated polyglycolized glycerides and/or ethanol is no greater than 20%, such as 1 -15%, 1 -10% or 1 -5% by weight.
In certain embodiments, the formulation comprises a triglyceride, a propylene glycol fatty acid ester, a polyethylene glycol sorbitan fatty acid ester, a saturated polyglycolized glyceride and an alcohol. Typically, the triglyceride is a long chain triglyceride, the propylene glycol fatty acid ester is propylene glycol caprylate/caprate, the polyethylene glycol sorbitan fatty acid ester is polyoxyethylated sorbitan monooleate, the saturated polyglycolized glyceride is lauryol macrogol glycerides and the alcohol is ethanol. In certain such embodiments, the formulation contains 1 -15% (e.g., 5-10%) by weight of the triglyceride, 10-25% (e.g., 15-20%) by weight of the propylene glycol fatty acid ester, 25-45% (e.g. 30- 40%) by weight of the polyethylene glycol sorbitan fatty acid ester, 10-25% (e.g. 15- 20%) by weight of the saturated polyglycolized glyceride and 5-20% (e.g., 10-15%) by weight of the alcohol.
In certain embodiments, the formulation comprises resveratrol, a monoacyl phospholipid, a PEG and a dihydric alcohol. Typically, the PEG is PEG 300 and the dihydric alcohol is propylene glycol. In certain such embodiments, the formulation contains 50-70% (e.g., 55-65%) by weight of the monoacyl phospholipid, 10-25% (e.g., 15-20%) by weight of the PEG and 10-25% (e.g., 15-20%) by weight of the dihydric alcohol In certain embodiments, the formulation comprises resveratrol, a polyethoxylated fatty acid, an alcohol-oil transesterification product and an alcohol. Typically, the polyethoxylated fatty acid is polyethoxylated glycol hydroxystearate, the alcohol-oil transesterification product is D-alpha-tocopheryl PEG 1000 succinate and the alcohol is ethanol. In certain such embodiments, the formulation contains 30-50% (e.g., 35-45%) by weight of the polyethoxylated fatty acid, 1-15% (e.g., 3- 8%) by weight of the alcohol-oil transesterification product and 40-60% (e.g., 45- 55%) by weight, of the alcohol.
In certain embodiments, the formulation comprises resveratrol, a triglyceride, an alcohol-oil transesterification product, a monoglyceride and an alcohol. Typically, the triglyceride is a medium chain triglyceride, the alcohol-oil transesterification product is a polyethxoylated hydrogenated castor oil, the monoglyceride is an oleate and the alcohol is ethanol. In certain such embodiments, the formulation contains 15-30% (e.g., 20-25%) by weight of the triglyceride, 35- 55% (e.g., 40-50%) by weight of the alcohol-oil transesterification product, 1-15% (e.g., 3-8%) by weight of the monoglyceride and 15-30% (e.g., 20-25%) by weight of the alcohol. In certain embodiments, the formulation comprises a polymer and a surfactant. Suitable polymers include polymers that have a full or partial positive charge, such as amine-containing polymers. An example of such a polymer is polyvinylpyrrolidone and derivatives thereof. Other examples include polyallylamine and polyvinylamine and derivatives thereof. Saturated polyglycolized glycerides are triglycerides that have been partially hydrolyzed with PEG, thereby forming a mixture of monoglycerides, diglycerides, triglycerides, fatty acid monoesters of PEG and fatty acid diesters of PEG. Examples of such compositions include Gelucire 44/14, Gelucire 50/13 and Gelucire 53/10. Monoglycerides are fatty acid monoesters of glycerol. Examples of monoglycerides include glyceryl monooleate, glyceryl 15 ricinoleate, glyceryl laurate, glyceryl monooleate, glyceryl caprylate/caprate, caprylic acid monoglycerides, and monoacetylated monoglycerides.
Triglycerides are fatty acid triesters of glycerol. Examples of triglycerides include Aceituno oil, Almond oil, Super Refined Almond Oil, Araehis oil, Babassu oil, Blackcurrant seed oil, Borage oil, Buffalo ground oil, Candlenut oil, Canola oil, Lipex 108, Castor oil, Chinese vegetable tallow oil, Cocoa butter, Coconut oil, Pureco 76, Coffee seed oil, Corn oil, Super Refined Corn Oil, Cottonseed oil, Super Refined Cottonseed Oil, Crambe oil, Cuphea species oil, Evening primrose oil, Grapeseed oil, Groundnut oil, Hemp seed oil, Illipe butter, Kapok seed oil, Linseed oil, Menhaden oil, Super Refined Menhaden Oil, Mowrah butter, Mustard seed oil, Oiticica oil, Olive oil, Super Refined Olive Oil, Palm oil, Palm kernel oil, Peanut oil, Super Refined Peanut Oil, Poppy seed oil, Rapeseed oil, Rice bran oil, Safflower oil, Super Refined Safflower Oil, Sal fat, Sesame oil, Super Refined Sesame Oil, Shark liver oil, Super Refined Shark Liver Oil, Shea nut oil, Soybean oil, Super Refined Soybean Oil, Stillingia oil, Sunflower oil, Tall oil, Tea seed oil, Tobacco seed oil, Tung oil, Ucuhuba Vernonia oil, Wheat germ oil, Super Refined Wheat Germ Oil, Hydrogenated Castorwax castor oil, Hydrogenated coconut oil, Hydrogenated cottonseed oil, Hydrogenated palm oil, Hydrogenated soybean oil, Hydrogenated vegetable oil, Hydrogenated cottonseed and castor oil, Partially hydrogenated soybean oil, Partially soy and cottonseed oil, Glyceryl tributyrate, Glyceryl tricaproate, Glyceryl tricaprylate, Glyceryl tricaprate, Glyceryl triundecanoate, Glyceryl trilaurate, Glyceryl trimyristate, Glyceryl tripalmitate, Glyceryl tristearate, Glyceryl triarchidate, Glyceryl trimyristoleate, Glyceryl tripalmitoleate, Glyceryl trioleate, Glyceryl trilinoleate, Glyceryl trilinolenate, Glyceryl tricaprylate/caprate, Glyceryl tricaprylate/caprate/laurate, Glyceryl tricaprylate/caprate/linoleate Glyceryl tricaprylate/caprate/stearate, Glyceryl tricaprylate/laurate/stearate, Glyceryl 1 ,2- caprylate-3-linoleate, Glyceryl l ,2-caprate-3-stearate, Glyceryl l ,2-laurate-3- myristate, Glyceryl l ,2-myristate-3-laurate, Glyceryl l ,3-palmitate-2-butyrate, Glyceryl l ,3-stearate-2-caprate, and Glyceryl l ,2-linoleate-3-caprylate. Fractionated triglycerides, modified triglycerides, synthetic triglycerides, and mixtures of triglycerides are considered triglycerides for this invention. A medium-chain triglyceride typically has 8 to 10 carbon atoms, whereas a long-chain triglyceride has 12 or more carbon atoms (typically 12-14, such as more than 16 carbon atoms is referred to as a very long chain). Diglycerides have only two fatty acid moieties attached to glycerol.
Monoacyl phospholipids include a hydrocarbon chain portion and a phospholipid portion. The hydrocarbon chain can either be unsaturated or saturated and can have between 10 to 24, preferably 14 to 18 carbon atoms. The lipids may be derived from natural plant, or animal or microbiological sources (e.g., phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylinositol (PI), phosphatidic acid (PI) and/or phosphatidylserine (PS)), synthesized or partially synthesized, including polyethyleneglycol (PEG) derived-monoacyl phospholipids (e.g., pegylated monoacyl phosphatidyl ethanolamine). Exemplary monoacyl phospholipids include enzyme modified (Phospholipase A2) soy PC, Egg PC, 1 - palmitoyl PC, 1 oleoyl PC, and 1 -stearoyl PC.
Examples of propylene glycol fatty acid esters include Propylene glycol monocaprylate, Propylene glycol monolaurate, Propylene glycol oleate, Propylene glycol myristate, Propylene glycol monostearate, Propylene glycol hydroxy stearate, Propylene glycol ricinoleate, Propylene glycol isostearate, Propylene glycol monooleate, Propylene glycol dicaprylate/dicaprate, Propylene glycol dioctanoate, Propylene glycol caprylate/caprate, Propylene glycol dilaurate, Propylene glycol distearate, Propylene glycol dicaprylate, and Propylene glycol dicaprate.
Examples of polyethylene glycol sorbitan fatty acid esters include PEG-10 sorbitan laurate, PEG-20 sorbitan monolaurate, PEG-4 sorbitan monolaurate, PEG- 80 sorbitan monolaurate, PEG-6 sorbitan monolaurate, PEG-20 sorbitan monopalmitate, PEG-20 sorbitan monostearate, PEG-4 sorbitan monostearate, PEG- 8 sorbitan monostearate, PEG-6 sorbitan monostearate, PEG-20 sorbitan tristearate, PEG-6 sorbitan tetrastearate, PEG-60 sorbitan tetrastearate, PEG-5 sorbitan monooleate, PEG-6 sorbitan monooleate, PEG-20 sorbitan monooleate, PEG-40 sorbitan oleate, PEG-20 sorbitan trioleate, PEG-6 sorbitan tetraoleate, PEG-30 sorbitan tetraoleate, PEG-40 sorbitan tetraoleate, PEG-20 sorbitan monoisostearate, PEG sorbitol hexaoleate and PEG-6 sorbitol hexastearate.
Examples of monohydric alcohols include methanol, ethanol, isopropanol and n-propanol. Typically, the alcohol is ethanol. Dihydric alcohols include two hydroxyl groups, such as ethylene glycol and propylene glycol.
Polyethylene glycols can have any suitable length, such as PEG- 100, PEG- 200, PEG-300, PEG-400, PEG-500, PEG- 1000 and so forth.
Examples of polyethoxylated fatty acids include PEG 4- 100 monolaurate, PEG 4- 100 monooleate, PEG 4-100 monostearate, PEG 400 distearate, PEG
100,200,300 monolaurate, PEG 100,200,300 monooleate, PEG 400 dioleate, PEG 400- 1000 monostearate, PEG-I stearate, PEG-2 stearate, PEG-2 oleate, PEG-4 laurate, PEG-4 oleate, PEG-4 stearate, PEG-5 stearate, PEG-5 oleate, PEG-6 oleate, PEG-7 oleate, PEG-6 laurate, PEG-7 laurate, PEG-6 stearate, PEG-8 laurate, PEG-8 oleate, PEG-8 stearate, PEG-9 oleate, PEG-9 stearate, PEG-10 laurate, PEG-10 oleate, PEG-10 stearate, PEG- 12 laurate, PEG- 12 oleate, PEG- 12 ricinoleate, PEG- 12 stearate, PEG- 15 stearate, PEG- 15 oleate, PEG-20 laurate, PEG-20 oleate, PEG- 20 stearate, PEG-25 stearate, PEG-32 laurate, PEG-32 oleate, PEG-32 stearate, PEG-30 stearate, PEG-40 laurate, PEG-40 oleate, PEG-40 stearate, PEG-45 stearate, PEG-50 stearate, PEG-55 stearate, PEG-100 oleate, PEG-100 stearate, PEG-200 oleate, PEG-400 oleate and PEG-600 oleate. A large number of surfactants of different degrees of hydrophobicity or hydrophilicity can be prepared by reaction of alcohols or polyalcohols with a variety of natural and/or hydrogenated oils. Most commonly, the oils used are castor oil or hydrogenated castor oil, or an edible vegetable oil such as com oil, olive oil, peanut oil, palm kernel oil, apricot kernel oil, or almond oil. Suitable alcohols include alcohols include glycerol, propylene glycol, ethylene glycol, polyethylene glycol, sorbitol, and pentaerythritol. Examples of alcohol-oil transesterification products include PEG-3 castor oil, PEG-5, 9, and 16 castor oil, PEG-20 castor oil, PEG-23 castor oil, PEG-30 castor oil, PEG-35 castor oil, PEG-38 castor oil, PEG-40 castor oil, PEG-50 castor oil, PEG-56 castor oil, PEG-60 castor oil, PEG-100 castor oil, PEG-200 castor oil, PEG-5 hydrogenated castor oil, PEG-7 hydrogenated castor oil, PEG- 10 hydrogenated castor oil, PEG-20 hydrogenated castor oil, PEG-25 hydrogenated castor oil, PEG-30 hydrogenated castor oil, PEG-40 hydrogenated castor oil, PEG-45 hydrogenated castor oil, PEG-50 hydrogenated castor oil, PEG- 60 hydrogenated castor oil, PEG-80 hydrogenated castor oil, PEG-100 hydrogenated castor oil, PEG-6 corn oil, PEG-6 almond oil, PEG-6 apricot kernel oil, PEG-6 olive oil, PEG-6 peanut oil, PEG-6 hydrogenated palm kernel oil, PEG-6 palm kernel oil, PEG-6 triolein, PEG-8 corn oil, PEG-20 corn glycerides, PEG-20 almond glycerides, PEG-25 trioleate, PEG-40 palm kernel oil, PEG-60 corn glycerides, PEG-60 almond glycerides, PEG-4 caprylic/ capric triglyceride, PEG-8 caprylic/capric glycerides, PEG-6 caprylic/capric glycerides, Lauroyl macrogol glyceride, Stearoyl macrogol glyceride, Mono, di, tri, tetra esters of vegetable oils and sorbitol, Pentaerythrityl tetraisostearate, Pentaerythrityl distearate, Pentaerythrityl tetraoleate, Pentaerythrityl tetrastearate, Pentaerythrityl tetracaprylate/tetracaprate and Pentaerythrityl tetraoctanoate. Also included as oils in this category of surfactants are oil-soluble vitamins, such as vitamins A, D, E, K, etc. Thus, derivatives of these vitamins, such as tocopheryl PEG-1000 succinate (TPGS, available from Eastman), are also included.
3. Exemplary Uses
In certain aspects, the invention provides methods for modulating the level and/or activity of a sirtuin protein and methods of use thereof. In certain embodiments, the invention provides methods for using formulations of the invention wherein the formulations activate a sirtuin protein, e.g., increase the level and/or activity of a sirtuin protein. Formulations that increase the level and/or activity of a sirtuin protein may be useful for a variety of therapeutic applications including, for example, increasing the lifespan of a cell, and treating and/or preventing a wide variety of diseases and disorders including, for example, diseases or disorders related to aging or stress, diabetes, obesity, neurodegenerative diseases, cardiovascular disease, blood clotting disorders, inflammation, cancer, and/or flushing, etc. The methods comprise administering to a subject in need thereof a pharmaceutically effective amount of a formulation. In certain embodiments, the formulations described herein may be taken alone or in combination with other compounds. In one embodiment, a mixture of resveratrol and one or more other sirtuin-modulating compounds may be administered to a subject in need thereof. In another embodiment, a formulation may be administered with one or more of the following compounds: butein, fisetin, piceatannol, or quercetin. In an exemplary embodiment, a formulation may be administered in combination with nicotinic acid. In yet another embodiment, formulations may be administered with one or more therapeutic agents for the treatment or prevention of various diseases, including, for example, cancer, diabetes, neurodegenerative diseases, cardiovascular disease, blood clotting, inflammation, flushing, obesity, ageing, stress, etc. In various embodiments, combination therapies comprising a formulation of the invention may refer to ( 1 ) pharmaceutical compositions that comprise one or more formulations in combination with one or more therapeutic agents (e.g., one or more therapeutic agents described herein); and (2) co-administration of one or more formulations with one or more therapeutic agents wherein the formulation of the invention and therapeutic agent have not been formulated in the same compositions (but may be present within the same kit or package, such as a blister pack or other multi-chamber package; connected, separately sealed containers (e.g., foil pouches) that can be separated by the user; or a kit where the formulation of the invention and other therapeutic agent(s) are in separate vessels). When using separate formulations, the formulation of the invention may be administered at the same, intermittent, staggered, prior to, subsequent to, or combinations thereof, with the administration of another therapeutic agent.
In certain embodiments, methods for reducing, preventing or treating diseases or disorders using a formulation of the invention may also comprise increasing the protein level of a sirtuin, such as human SIRTl , SIRT2 and/or SIRT3, or homologs thereof. Increasing protein levels can be achieved by introducing into a cell one or more copies of a nucleic acid that encodes a sirtuin. For example, the level of a sirtuin can be increased in a mammalian cell by introducing into the mammalian cell a nucleic acid encoding the sirtuin, e.g., increasing the level of SIRTl by introducing a nucleic acid encoding the amino acid sequence set forth in GenBank Accession No. NP_036370 and/or increasing the level of SIRT3 by introducing a nucleic acid encoding the amino acid sequence set forth in GenBank Accession No. AAH01042. The nucleic acid may be under the control of a promoter that regulates the expression of the SIRTl and/or SIRT3 nucleic acid. Alternatively, the nucleic acid may be introduced into the cell at a location in the genome that is downstream of a promoter. Methods for increasing the level of a protein using these methods are well known in the art.
A nucleic acid that is introduced into a cell to increase the protein level of a sirtuin may encode a protein that is at least about 80%, 85%, 90%, 95%, 98%, or 99% identical to the sequence of a sirtuin, e.g., SIRTl (GenBank Accession No. NP 036370) and/or SIRT3 (GenBank Accession No. AAHO 1042) protein. For example, the nucleic acid encoding the protein may be at least about 80%, 85%, 90%, 95%, 98%, or 99% identical to a nucleic acid encoding a SIRTl (e.g. GenBank Accession No. NM_012238) and/or SIRT3 (e.g., GenBank Accession No. BCOO 1042) protein. The nucleic acid may also be a nucleic acid that hybridizes, preferably under stringent hybridization conditions, to a nucleic acid encoding a wild-type sirtuin, e.g., SIRTl (GenBank Accession No. NM_012238) and/or SIRT3 (e.g., GenBank Accession No. BCOOl 042) protein. Stringent hybridization conditions may include hybridization and a wash in 0.2 x SSC at 65 0C. When using a nucleic acid that encodes a protein that is different from a wild-type sirtuin protein, such as a protein that is a fragment of a wild-type sirtuin, the protein is preferably biologically active, e.g., is capable of deacetylation. It is only necessary to express in a cell a portion of the sirtuin that is biologically active. For example, a protein that differs from wild-type SIRTl having GenBank Accession No. NP 036370, preferably contains the core structure thereof. The core structure sometimes refers to amino acids 62-293 of GenBank Accession No. NP 036370, which are encoded by nucleotides 237 to 932 of GenBank Accession No. NM Ol 2238, which encompasses the NAD binding as well as the substrate binding domains. The core domain of SIRTl may also refer to about amino acids 261 to 447 of GenBank Accession No. NP 036370, which are encoded by nucleotides 834 to 1394 of GenBank Accession No. NM 012238; to about amino acids 242 to 493 of GenBank Accession No. NP 036370, which are encoded by nucleotides 777 to 1532 of GenBank Accession No. NM 012238; or to about amino acids 254 to 495 of
GenBank Accession No. NP 036370, which are encoded by nucleotides 813 to 1538 of GenBank Accession No. NM_012238. Whether a protein retains a biological function, e.g., deacetylation capabilities, can be determined according to methods known in the art. In certain embodiments, methods for reducing, preventing or treating diseases or disorders using a formulation of the invention may also comprise decreasing the protein level of a sirtuin, such as human SIRTl , SIRT2 and/or SIRT3, or homologs thereof. Decreasing a sirtuin protein level can be achieved according to methods known in the art. For example, an siRNA, an antisense nucleic acid, or a ribozyme targeted to the sirtuin can be expressed in the cell. A dominant negative sirtuin mutant, e.g., a mutant that is not capable of deacetylating, may also be used. For example, mutant H363Y of SIRTl , described, e.g., in Luo et al. (2001 ) Cell 107: 137 can be used. Alternatively, agents that inhibit transcription can be used.
Methods for modulating sirtuin protein levels also include methods for modulating the transcription of genes encoding sirtuins, methods for stabilizing/destabilizing the corresponding mRNAs, and other methods known in the art. Aging/Stress
In one embodiment, the invention provides a method extending the lifespan of a cell, extending the proliferative capacity of a cell, slowing ageing of a cell, promoting the survival of a cell, delaying cellular senescence in a cell, mimicking the effects of calorie restriction, increasing the resistance of a cell to stress, or preventing apoptosis of a cell, by contacting the cell with a formulation of the invention that increases the level and/or activity of a sirtuin protein.
The methods described herein may be used to increase the amount of time that cells, particularly primary cells (i.e., cells obtained from an organism, e.g., a human), may be kept alive in a cell culture. Embryonic stem (ES) cells and pluripotent cells, and cells differentiated therefrom, may also be treated with a - formulation of the invention to keep the cells, or progeny thereof, in culture for longer periods of time. Such cells can also be used for transplantation into a subject, e.g., after ex vivo modification.
In one embodiment, cells that are intended to be preserved for long periods of time may be treated with a formulation of the invention. The cells may be in suspension (e.g.. blood cells, serum, biological growth media, etc.) or in tissues or organs. For example, blood collected from an individual for purposes of transfusion may be treated with a formulation of the invention to preserve the blood cells for longer periods of time. Additionally, blood to be used for forensic purposes may also be preserved using a formulation of the invention. Other cells that may be treated to extend their lifespan or protect against apoptosis include cells for consumption, e.g., cells from non-human mammals (such as meat) or plant cells (such as vegetables). Formulations of the invention may also be applied during developmental and growth phases in mammals, plants, insects or microorganisms, in order to, e.g., alter, retard or accelerate the developmental and/or growth process.
In another embodiment, formulations of the invention may be used to treat cells useful for transplantation or cell therapy, including, for example, solid tissue grafts, organ transplants, cell suspensions, stem cells, bone marrow cells, etc. The cells or tissue may be an autograft, an allograft, a syngraft or a xenograft. The cells or tissue may be treated with the formulation of the invention prior to administration/implantation, concurrently with administration/implantation, and/or post administration/implantation into a subject. The cells or tissue may be treated prior to removal of the cells from the donor individual, ex vivo after removal of the cells or tissue from the donor individual, or post implantation into the recipient. For example, the donor or recipient individual may be treated systemically with a formulation of the invention or may have a subset of cells/tissue treated locally with a formulation of the invention. In certain embodiments, the cells or tissue (or donor/recipient individuals) may additionally be treated with another therapeutic agent useful for prolonging graft survival, such as, for example, an immunosuppressive agent, a cytokine, an angiogenic factor, etc.
In yet other embodiments, cells may be treated with a formulation of the invention in vivo, e.g., to increase their lifespan or prevent apoptosis. For example, skin can be protected from aging (e.g., developing wrinkles, loss of elasticity, etc.) by treating skin or epithelial cells with a formulation of the invention. In an exemplary embodiment, skin is contacted with a formulation of the invention. Exemplary skin afflictions or skin conditions that may be treated in accordance with the methods described herein include disorders or diseases associated with or caused by inflammation, sun damage or natural aging. For example, the compositions find utility in the prevention or treatment of contact dermatitis (including irritant contact dermatitis and allergic contact dermatitis), atopic dermatitis (also known as allergic eczema), actinic keratosis, keratinization disorders (including eczema), epidermolysis bullosa diseases (including penfigus), exfoliative dermatitis, seborrheic dermatitis, erythemas (including erythema multiforme and erythema nodosum), damage caused by the sun or other light sources, discoid lupus erythematosus, dermatomyositis, psoriasis, skin cancer and the effects of natural aging. In another embodiment, a formulation of the invention may be used for the treatment of wounds and/or burns to promote healing, including, for example, first-, second- or third-degree burns and/or thermal, chemical or electrical burns. The formulations may be administered topically, to the skin or mucosal tissue, as an ointment, lotion, cream, microemulsion, gel, solution or the like, as further described herein, within the context of a dosing regimen effective to bring about the desired result.
Topical formulations may also be used as preventive, e.g., chemopreventive, compositions. When used in a chemopreventive method, susceptible skin is treated prior to any visible condition in a particular individual.
Formulations of the invention may be delivered locally or systemically to a subject. In one embodiment, a formulation of the invention is delivered locally to a tissue or organ of a subject by injection, topical formulation, etc. In another embodiment, a formulation of the invention may be used for treating or preventing a disease or condition induced or exacerbated by cellular senescence in a subject; methods for decreasing the rate of senescence of a subject, e.g., after onset of senescence; methods for extending the lifespan of a subject; methods for treating or preventing a disease or condition relating to lifespan; methods for treating or preventing a disease or condition relating to the proliferative capacity of cells; and methods for treating or preventing a disease or condition resulting from cell damage or death. In certain embodiments, the method does not act by decreasing the rate of occurrence of diseases that shorten the lifespan of a subject. In certain embodiments, a method does not act by reducing the lethality caused by a disease, such as cancer.
In yet another embodiment, a formulation of the invention may be administered to a subject in order to generally increase the lifespan of its cells and to protect its cells against stress and/or against apoptosis. It is believed that treating a subject with a compound described herein is similar to subjecting the subject to hoπnesis, i.e., mild stress that is beneficial to organisms and may extend their lifespan. Formulations of the invention may be administered to a subject to prevent aging and aging-related consequences or diseases, such as stroke, heart disease, heart failure, arthritis, high blood pressure, and Alzheimer's disease. Other conditions that can be treated include ocular disorders, e.g., associated with the aging of the eye, such as cataracts, glaucoma, and macular degeneration.
Formulations of the invention can also be administered to subjects for treatment of diseases, e.g., chronic diseases, associated with cell death, in order to protect the cells from cell death. Exemplary diseases include those associated with neural cell death, neuronal dysfunction, or muscular cell death or dysfunction, such as Parkinson's disease, Alzheimer's disease, multiple sclerosis, amniotropic lateral sclerosis, and muscular dystrophy; AIDS; fulminant hepatitis; diseases linked to degeneration of the brain, such as Creutzfeld-Jakob disease, retinitis pigmentosa and cerebellar degeneration; myelodysplasis such as aplastic anemia; ischemic diseases such as myocardial infarction and stroke; hepatic diseases such as alcoholic hepatitis, hepatitis B and hepatitis C; joint diseases such as osteoarthritis; atherosclerosis; alopecia; damage to the skin due to UV light; lichen planus; atrophy of the skin; cataract; and graft rejections. Cell death can also be caused by surgery, drug therapy, chemical exposure or radiation exposure.
Formulation of the inventions can also be administered to a subject suffering from an acute disease, e.g., damage to an organ or tissue, e.g., a subject suffering from stroke or myocardial infarction or a subject suffering from a spinal cord injury. Formulations of the invention may also be used to repair an alcoholic's liver. Car dio vascular Disease In another embodiment, the invention provides a method for treating and/or preventing a cardiovascular disease by administering to a subject in need thereof a formulation of the invention.
Cardiovascular diseases that can be treated or prevented using the formulations of the invention include cardiomyopathy or myocarditis; such as idiopathic cardiomyopathy, metabolic cardiomyopathy, alcoholic cardiomyopathy, drug-induced cardiomyopathy, ischemic cardiomyopathy, and hypertensive cardiomyopathy. Also treatable or preventable using a formulation of the invention are atheromatous disorders of the major blood vessels (macrovascular disease) such as the aorta, the coronary arteries, the carotid arteries, the cerebrovascular arteries, the renal arteries, the iliac arteries, the femoral arteries, and the popliteal arteries. Other vascular diseases that can be treated or prevented include those related to platelet aggregation, the retinal arterioles, the glomerular arterioles, the vasa nervorum, cardiac arterioles, and associated capillary beds of the eye, the kidney, the heart, and the central and peripheral nervous systems. The formulations of the invention may also be used for increasing HDL levels in plasma of an individual. Yet other disorders that may be treated with a formulation of the invention include restenosis, e.g., following coronary intervention, and disorders relating to an abnormal level of high density and low density cholesterol.
In one embodiment, a formulation of the invention may be administered as part of a combination therapeutic with another cardiovascular agent including, for example, an anti-arrhythmic agent, an antihypertensive agent, a calcium channel blocker, a cardioplegic solution, a cardiotonic agent, a fibrinolytic agent, a sclerosing solution, a vasoconstrictor agent, a vasodilator agent, a nitric oxide donor, a potassium channel blocker, a sodium channel blocker, statins, or a natriuretic agent.
In one embodiment, a formulation of the invention may be administered as part of a combination therapeutic with an anti-arrhythmia agent. Anti-arrhythmia agents are often organized into four main groups according to their mechanism of _. action: type I, sodium channel blockade; type II, beta-adrenergic blockade; type 111, repolarization prolongation; and type IV, calcium channel blockade. Type I antiarrhythmic agents include lidocaine, moricizine, mexiletine, tocainide, procainamide, encainide, flecanide, tocainide, phenytoin, propafenone, quinidine, disopyramide, and flecainide. Type II anti-arrhythmic agents include propranolol and esmolol. Type III includes agents that act by prolonging the duration of the action potential, such as amiodarone, artilide, bretylium, clofilium, isobutilide, sotalol, azimilide, dofetilide, dronedarone, ersentilide, ibutilide, tedisamil, and trecetilide. Type IV anti-arrhythmic agents include verapamil, diltaizem, digitalis, adenosine, nickel chloride, and magnesium ions. In another embodiment, a formulation of the invention may be administered as part of a combination therapeutic with another cardiovascular agent. Examples of cardiovascular agents include vasodilators, for example, hydralazine; angiotensin converting enzyme inhibitors, for example, captopril; anti-anginal agents, for example, isosorbide nitrate, glyceryl trinitrate and pentaerythritol tetranitrate; antiarrhythmic agents, for example, quinidine, procainaltide and lignocaine; cardioglycosides, for example, digoxin and digitoxin; calcium antagonists, for example, verapamil and nifedipine; diuretics, such as thiazides and related compounds, for example, bendrofluazide, chlorothiazide, chlorothalidone, hydrochlorothiazide and other diuretics, for example, fursemide and triamterene, and sedatives, for example, nitrazepam, flurazepam and diazepam.
Other exemplary cardiovascular agents include, for example, a cyclooxygenase inhibitor such as aspirin or indomethacin, a platelet aggregation inhibitor such as clopidogrel, ticlopidene or aspirin, fibrinogen antagonists or a diuretic such as chlorothiazide, hydrochlorothiazide, flumethiazide, hydroflumethiazide, bendro flumethiazide, methyl chlorthiazide, trichloromethiazide, polythiazide or benzthiazide as well as ethacrynic acid tricrynafen, chlorthalidone, furosemide, musolimine, bumetanide, triamterene, amiloride and spironolactone and salts of such compounds, angiotensin converting enzyme inhibitors such as captopril, zofenopril, fosinopril, enalapril, ceranopril, cilazopril, delapril, pentopril, quinapril, ramipril, lisinopril, and salts of such compounds, angiotensin II antagonists such as losartan, irbesartan or valsartan, thrombolytic agents such as tissue plasminogen activator (tPA), recombinant tPA, streptokinase, urokinase, prourokinase, and anisoylated plasminogen streptokinase activator complex (APSAC, Eminase, Beecham Laboratories), or animal salivary gland plasminogen activators, calcium channel blocking agents such as verapamil, nifedipine or diltiazem, thromboxane receptor antagonists such as ifetroban, prostacyclin mimetics, or phosphodiesterase inhibitors. Such combination products if formulated as a fixed dose employ the compounds of this invention within the dose range described above and the other pharmaceutically active agent within its approved dose range. Yet other exemplary cardiovascular agents include, for example, vasodilators, e.g., bencyclane, cinnarizine, citicoline, cyclandelate, cyclonicate, ebumamonine, phenoxezyl, flunarizine, ibudilast, ifenprodil, lomerizine, naphlole, nikamate, nosergoline, nimodipine, papaverine, pentifylline, nofedoline, vincamin, vinpocetine, vichizyl, pentoxifylline, prostacyclin derivatives (such as prostaglandin El and prostaglandin 12), an endothelin receptor blocking drug (such as bosentan), diltiazem, nicorandil, and nitroglycerin. Examples of the cerebral protecting drug include radical scavengers (such as edaravone, vitamin E, and vitamin C), glutamate antagonists, AMPA antagonists, kainate antagonists, NMDA antagonists, GABA agonists, growth factors, opioid antagonists, phosphatidylcholine precursors, serotonin agonists, Na+/Ca2+ channel inhibitory drugs, and K+ channel opening drugs. Examples of the brain metabolic stimulants include amantadine, tiapride, and gamma-aminobutyric acid. Examples of the anticoagulant include heparins (such as heparin sodium, heparin potassium, dalteparin sodium, dalteparin calcium, heparin calcium, parnaparin sodium, reviparin sodium, and danaparoid sodium), warfarin, enoxaparin, argatroban, batroxobin, and sodium citrate. Examples of the antiplatelet drug include ticlopidine hydrochloride, dipyridamole, cilostazol, ethyl icosapentate, sarpogrelate hydrochloride, dilazep hydrochloride, trapidil, a nonsteroidal antiinflammatory agent (such as aspirin), beraprostsodium, iloprost, and indobufene. Examples of the thrombolytic drug include urokinase, tissue-type plasminogen activators (such as alteplase, tisokinase, nateplase, pamiteplase, monteplase, and rateplase), and nasaruplase. Examples of the antihypertensive drug include angiotensin converting enzyme inhibitors (such as captopril, alacepril, lisinopril, imidapril, quinapril, temocapril, delapril, benazepril, cilazapril, trandolapril, enalapril, ceronapril, fosinopril, imadapril, mobertpril, perindopril, ramipril, spirapril, and randolapril), angiotensin 11 antagonists (such as losartan, candesartan, valsartan, eprosartan, and irbesartan), calcium channel blocking drugs (such as aranidipine, efonidipine, nicardipine, bamidipine, benidipine, manidipine, cilnidipine, nisoldipine, nitrendipine, nifedipine, nilvadipine, felodipine, amlodipine, diltiazem, bepridil, clentiazem, phendilin, galopamil, mibefradil, prenylamine, semotiadil, terodiline, verapamil, cilnidipine, elgodipine, isradipine, lacidipine, lercanidipine, nimodipine, cinnarizine, flunarizine, lidoflazine, lomerizine, bencyclane, etafenone, and perhexiline), β-adrenaline receptor blocking drugs (propranolol, pindolol, indenolol, carteolol, bunitrolol, atenolol, acebutolol, metoprolol, timolol, nipradilol, penbutolol, nadolol, tilisolol, carvedilol, bisoprolol, betaxolol, celiprolol, bopindolol, bevantolol, labetalol, alprenolol, amosulalol, arotinolol, befunolol, bucumolol, bufetolol, buferalol, buprandolol, butylidine, butofϊlolol, carazolol, cetamolol, cloranolol, dilevalol, epanolol, levobunolol, mepindolol, metipranolol, moprolol, nadoxolol, nevibolol, oxprenolol, practol, pronetalol, sotalol, sufinalol, talindolol, tertalol, toliprolol, xybenolol, and esmolol), α-receptor blocking drugs (such as amosulalol, prazosin, terazosin, doxazosin, bunazosin, urapidil, phentolamine, arotinolol, dapiprazole, fenspiride, indoramin, labetalol, naftopidil, nicergoline, tamsulosin, tolazoline, trimazosin, and yohimbine), sympathetic nerve inhibitors (such as clonidine, guanfacine, guanabenz, methyldopa, and reserpine), hydralazine, todralazine, budralazine, and cadralazine. Examples of the antianginal drug include nitrate drugs (such as amyl nitrite, nitroglycerin, and isosorbide), β-adrenaline receptor blocking drugs (such as propranolol, pindolol, indenolol, carteolol, bunitrolol, atenolol, acebutolol, metoprolol, timolol, nipradilol, penbutolol, nadolol, tilisolol, carvedilol, bisoprolol, betaxolol, celiprolol, bopindolol, bevantolol, labetalol, alprenolol, amosulalol, arotinolol, befunolol, bucumolol, bufetolol, buferalol, buprandolol, butylidine, butofilolol, carazolol, cetamolol, cloranolol, dilevalol, epanolol, levobunolol, mepindolol, metipranolol, moprolol, nadoxolol, nevibolol, oxprenolol, practol, pronetalol, sotalol, sufinalol, talindolol, tertalol, toliprolol, andxybenolol), calcium channel blocking drugs (such as aranidipine, efonidipine, nicardipine, bamidipine, benidipine, manidipine, cilnidipine, nisoldipine, nitrendipine, nifedipine, nilvadipine, felodipine, amlodipine, diltiazem, bepridil, clentiazem, phendiline, galopamil, mibefradil, prenylamine, semotiadil, terodiline, verapamil, cilnidipine, elgodipine, isradipine, lacidipine, lercanidipine, nimodipine, cinnarizine, flunarizine, lidoflazine, lomerizine, bencyclane, etafenone, and perhexiline) trimetazidine, dipyridamole, etafenone, dilazep, trapidil, nicorandil, enoxaparin, and aspirin. Examples of the diuretic include thiazide diuretics (such as hydrochlorothiazide, methyclothiazide, trichlormethiazide, benzylhydrochlorothiazide, and penflutizide), loop diuretics (such as furosemide, etacrynic acid, bumetanide, piretanide, azosemide, and torasemide), K+ sparing diuretics (spironolactone, triamterene, andpotassiumcanrenoate), osmotic diuretics (such as isosorbide, D-mannitol, and glycerin), nonthiazide diuretics (such as meticrane, tripamide, chlorthalidone, and mefruside), and acetazolamide. Examples of the cardiotonic include digitalis formulations (such as digitoxin, digoxin, methyldigoxin, deslanoside, vesnarinone, lanatoside C, and proscillaridin), xanthine formulations (such as aminophylline, choline theophylline, diprophylline, and proxyphylline), catecholamine formulations (such as dopamine, dobutamine, and docarpamine), PDE III inhibitors (such as amrinone, olprinone, and milrinone), denopamine, ubidecarenone, pimobendan, levosimendan, aminoethylsulfonic acid, vesnarinone, carperitide, and colforsin daropate. Examples of the antiarrhythmic drug include ajmaline, pirmenol, procainamide, cibenzoline, disopyramide, quinidine, aprindine, mexiletine, lidocaine, phenyloin, pilsicainide, propafenone, flecainide, atenolol, acebutolol, sotalol, propranolol, metoprolol, pindolol, amiodarone, nifekalant, diltiazem, bepridil, and verapamil. Examples of the antihyperlipidemic drug include atorvastatin, simvastatin, pravastatin sodium, fluvastatin sodium, clinofibrate, clofibrate, simfibrate, fenofibrate, bezafibrate, colestimide, and colestyramine. Examples of the immunosuppressant include azathioprine, mizoribine, cyclosporine, tacrolimus, gusperimus, and methotrexate. Cell Death/Cancer
Formulations of the invention may be administered to subjects who have recently received or are likely to receive a dose of radiation or toxin. In one embodiment, the dose of radiation or toxin is received as part of a work-related or medical procedure, e.g., working in a nuclear power plant, flying an airplane, an X- ray, CAT scan, or the administration of a radioactive dye for medical imaging; in such an embodiment, the formulation of the invention is administered as a prophylactic measure. In another embodiment, the radiation or toxin exposure is received unintentionally, e.g., as a result of an industrial accident, habitation in a location of natural radiation, terrorist act, or act of war involving radioactive or toxic material. In such a case, the formulation of the invention is preferably administered as soon as possible after the exposure to inhibit apoptosis and the subsequent development of acute radiation syndrome. Formulations of the invention may also be used for treating and/or preventing cancer. In certain embodiments, formulations of the invention may be used for treating and/or preventing cancer. Calorie restriction has been linked to a reduction in the incidence of age-related disorders including cancer (see e.g., Bordone and Guarente, Nat. Rev. MoI. Cell Biol. (2005 epub); Guarente and Picard, Cell 120: 473-82 (2005); Berrigan, et al., Carcinogenesis 23: 817-822 (2002); and Heilbronn and Ravussin, Am. J. Clin. Nutr. 78: 361 -369 (2003)). Additionally, the Sir2 protein from yeast has been- shown to be required for lifespan extension by glucose restriction (see e.g., Lin et al., Science 289: 2126-2128 (2000); Anderson et al., Nature 423: 181 -185 (2003)), a yeast model for calorie restriction. Accordingly, an increase in the level and/or activity of a sirtuin protein may be useful for treating and/or preventing the incidence of age-related disorders, such as, for example, cancer. Exemplary cancers that may be treated using a formulation of the invention are those of the brain and kidney; hormone-dependent cancers including breast, prostate, testicular, and ovarian cancers; lymphomas, and leukemias. In cancers associated with solid tumors, a formulation of the invention may be administered directly into the tumor. Cancer of blood cells, e.g., leukemia, can be treated by administering a modulating compound into the blood stream or into the bone marrow. Benign cell growth can also be treated, e.g., warts. Other diseases that can be treated include autoimmune diseases, e.g., systemic lupus erythematosus, scleroderma, and arthritis, in which autoimmune cells should be removed. Viral infections such as herpes, HIV, adenovirus, and HTLV-I associated malignant and benign disorders can also be treated by administration of a formulation of the invention. Alternatively, cells can be obtained from a subject, treated ex vivo to remove certain undesirable cells, e.g., cancer cells, and administered back to the same or a different subject.
Chemotherapeutic agents that may be coadministered with formulation of the invention as having anti-cancer activity (e.g., compounds that induce apoptosis, compounds that reduce lifespan or compounds that render cells sensitive to stress) include: aminoglutethimide, amsacrine, anastrozole, asparaginase, beg, bicalutamide, bleomycin, buserelin, busulfan, campothecin, capecitabine, carboplatin, carmustine, chlorambucil, cisplatin, cladribine, clodronate, colchicine, cyclophosphamide, cyproterone, cytarabine, dacarbazine, dactinomycin, daunorubicin, dienestrol, diethylstilbestrol, docetaxel, doxorubicin, epirubicin, estradiol, estramustine, etoposide, exemestane, filgrastim, fludarabine, fludrocortisone, fluorouracil, fluoxymesterone, flutamide, gemcitabine, genistein, goserelin, hydroxyurea, idarubicin, ifosfamide, imatinib, interferon, irinotecan, ironotecan, letrozole, leucovorin, leuprolide, levamisole, lomustine, mechlorethamine, medroxyprogesterone, megestrol, melphalan, mercaptopurine, mesna, methotrexate, mitomycin, mitotane, mitoxantrone, nilutamide, nocodazole, octreotide, oxaliplatin, paclitaxel, pamidronate, pentostatin, plicamycin, porfimer, procarbazine, raltitrexed, rituximab, streptozocin, suramin, tamoxifen, temozolomide, teniposide, testosterone, thioguanine, thiotepa, titanocene dichloride, topotecan, trastuzumab, tretinoin, vinblastine, vincristine, vindesine, and vinorelbine.
These chemotherapeutic agents may be categorized by their mechanism of action into, for example, following groups: anti-metabolites/anti-cancer agents, such as pyrimidine analogs (5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine) and purine analogs, folate antagonists and related inhibitors (mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine (cladribine)); antiproliferative/antimitotic agents including natural products such as vinca alkaloids (vinblastine, vincristine, and vinorelbine), microtubule disrupters such as taxane (paclitaxel, docetaxel), vincristin, vinblastin, nocodazole, epothilones and navelbine, epidipodophyllotoxins (teniposide), DNA damaging agents (actinomycin, amsacrine, anthracyclines, bleomycin, busulfan, camptothecin, carboplatin, chlorambucil, cisplatin, cyclophosphamide, Cytoxan, dactinomycin, daunorubicin, docetaxel, doxorubicin, epirubicin, hexamethylmelamineoxaliplatin, iphosphamide, melphalan, merchlorethamine, mitomycin, mitoxantrone, nitrosourea, paclitaxel, plicamycin, procarbazine, teniposide. triethylenethiophosphoramide and etoposide (VP 16)); antibiotics such as dactinomycin (actinomycin D), daunorubicin, doxorubicin (adriamycin), idarubicin, anthracyclines, mitoxantrone, bleomycins, plicamycin (mithramycin) and mitomycin; enzymes (L-asparaginase which systemically metabolizes L-asparagine and deprives cells which do not have the capacity to synthesize their own asparagine); antiplatelet agents; antiproliferative/antimitotic alkylating agents such as nitrogen mustards (mechlorethamine, cyclophosphamide and analogs, melphalan, chlorambucil), ethylenimines and methylmelamines (hexamethylmelamine and thiotepa), alkyl sulfonates-busulfan, nitrosoureas (carmustine (BCNU) and analogs, streptozocin), trazenes - dacarbazinine (DTIC); antiproliferative/antimitotic antimetabolites such as folic acid analogs (methotrexate); platinum coordination complexes (cisplatin, carboplatin), procarbazine, hydroxyurea, mitotane, aminoglutethimide; hormones, hormone analogs (estrogen, tamoxifen, goserelin, bicalutamide, nilutamide) and aromatase inhibitors (letrozole, anastrozole); anticoagulants (heparin, synthetic heparin salts and other inhibitors of thrombin); fibrinolytic agents (such as tissue plasminogen activator, streptokinase and urokinase), aspirin, COX-2 inhibitors, dipyridamole, ticlopidine, clopidogrel, abciximab; antimigratory agents; antisecretory agents (breveldin); immunosuppressives (cyclosporine, tacrolimus (FK-506), sirolimus (rapamycin), azathioprine, mycophenolate mofetil); anti- angiogenic compounds (TNP-470, genistein) and growth factor inhibitors (vascular endothelial growth factor (VEGF) inhibitors, fibroblast growth factor (FGF) - inhibitors, epidermal growth factor (EGF) inhibitors); angiotensin receptor blocker; nitric oxide donors; anti-sense oligonucleotides; antibodies (trastuzumab); cell cycle inhibitors and differentiation inducers (tretinoin); mTOR inhibitors, topoisomerase inhibitors (doxorubicin (adriamycin), amsacrine, camptothecin, daunorubicin, dactinomycin, eniposide, epirubicin, etoposide, idarubicin, irinotecan (CPT- 1 1 ) and mitoxantrone, topotecan, irinotecan), corticosteroids (cortisone, dexamethasone, hydrocortisone, methylpednisolone, prednisone, and prenisolone); growth factor signal transduction kinase inhibitors; mitochondrial dysfunction inducers and caspase activators; chromatin disruptors.
These chemotherapeutic agents may be used by themselves with a formulation of the invention. Many combinatorial therapies have been developed, including but not limited to those listed in Table 1.
Table 1 : Exemplary combinatorial therapies for the treatment of cancer.
In addition to conventional chemotherapeutics, a formulation of the invention can also be used with antisense RNA, RNAi or other polynucleotides to inhibit the expression of the cellular components that contribute to unwanted cellular proliferation that are targets of conventional chemotherapy. Such targets are, merely to illustrate, growth factors, growth factor receptors, cell cycle regulatory proteins, transcription factors, or signal transduction kinases.
Combination therapies comprising a formulation of the invention and a conventional chemotherapeutic agent may be advantageous over combination therapies known in the art because the combination allows the conventional chemotherapeutic agent to exert greater effect at lower dosage. In a preferred embodiment, the effective dose (ED50) for a chemotherapeutic agent, or combination of conventional chemotherapeutic agents, when used in combination with a formulation of the invention is at least 2 fold less than the ED50 for the chemotherapeutic agent alone, and even more preferably at 5 fold, 10 fold or even 25 fold less. Conversely, the therapeutic index (TI) for such chemotherapeutic agent or combination of such chemotherapeutic agent when used in combination with a formulation of the invention described herein can be at least 2 fold greater than the TI for conventional chemotherapeutic regimen alone, and even more preferably at 5 fold, 10 fold or even 25 fold greater. Neuronal Diseases/Disorders
In certain aspects, formulations of the invention can be used to treat patients suffering from neurodegenerative diseases, and traumatic or mechanical injury to the central nervous system (CNS), spinal cord or peripheral nervous system (PNS).
Neurodegenerative disease typically involves reductions in the mass and volume of the human brain, which may be due to the atrophy and/or death of brain cells, which are far more profound than those in a healthy person that are attributable to aging. Neurodegenerative diseases can evolve gradually, after a long period of normal brain function, due to progressive degeneration (e.g., nerve cell dysfunction and death) of specific brain regions. Alternatively, neurodegenerative diseases can have a quick onset, such as those associated with trauma or toxins. The actual onset of brain degeneration may precede clinical expression by many years. Examples of neurodegenerative diseases include, but are not limited to, Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS; Lou Gehrig's disease), diffuse Lewy body disease, chorea- acanthocytosis, primary lateral sclerosis, ocular diseases (ocular neuritis), chemotherapy-induced neuropathies (e.g., from vincristine, paclitaxel, bortezomib), diabetes-induced neuropathies and Friedreich's ataxia. A formulation of the invention can be used to treat these disorders and others as described below.
AD is a chronic, incurable, and unstoppable CNS disorder that occurs gradually, resulting in memory loss, unusual behavior, personality changes, and a decline in thinking abilities. These losses are related to the death of specific types of brain cells and the breakdown of connections and their supporting network (e.g. glial cells) between them. AD has been described as childhood development in reverse. In most people with AD, symptoms appear after the age 60. The earliest symptoms include loss of recent memory, faulty judgment, and changes in personality. Later in the disease, those with AD may forget how to do simple tasks like washing their hands. Eventually people with AD lose all reasoning abilities and become dependent on other people for their everyday care. Finally, the disease becomes so debilitating that patients are bedridden and typically develop coexisting illnesses. PD is a chronic, incurable, and unstoppable CNS disorder that occurs gradually and results in uncontrolled body movements, rigidity, tremor, and dyskinesia. These motor system problems are related to the death of brain cells in an area of the brain that produces dopamine, a chemical that helps control muscle activity. In most people with PD, symptoms appear after age 50. The initial symptoms of PD are a pronounced tremor affecting the extremities,- notably in the hands or lips. Subsequent characteristic symptoms of PD are stiffness or slowness of movement, a shuffling walk, stooped posture, and impaired balance. There are wide ranging secondary symptoms such as memory loss, dementia, depression, emotional changes, swallowing difficulties, abnormal speech, sexual dysfunction, and bladder and bowel problems. These symptoms will begin to interfere with routine activities, such as holding a fork or reading a newspaper. Finally, people with PD become so profoundly disabled that they are bedridden.
ALS (motor neuron disease) is a chronic, incurable, and unstoppable CNS disorder that attacks the motor neurons, components of the CNS that connect the brain to the skeletal muscles. In ALS, the motor neurons deteriorate and eventually die, and though a person's brain normally remains fully functioning and alert, the command to move never reaches the muscles. Most people who get ALS are between 40 and 70 years old. The first motor neurons that weaken are those controlling the arms or legs. Those with ALS may have trouble walking, they may drop things, fall, slur their speech, and laugh or cry uncontrollably. Eventually the muscles in the limbs begin to atrophy from disuse. This muscle weakness will become debilitating and a person will need a wheel chair or become unable to function out of bed.
The causes of these neurological diseases have remained largely unknown. They are conventionally defined as distinct diseases, yet clearly show extraordinary similarities in basic processes and commonly demonstrate overlapping symptoms far greater than would be expected by chance alone. Current disease definitions fail to properly deal with the issue of overlap and a new classification of the neurodegenerative disorders has been called for.
HD is another neurodegenerative disease resulting from genetically programmed degeneration of neurons in certain areas of the brain. This degeneration causes uncontrolled movements, loss of intellectual faculties, and emotional disturbance. HD is a familial disease, passed from parent to child through a dominant mutation in the wild-type gene. Some early symptoms of HD are mood swings, depression, irritability or trouble driving, learning new things, remembering a fact, or making a decision. As the disease progresses, concentration on intellectual tasks becomes increasingly difficult and the patient may have difficulty feeding himself or herself and swallowing.
Tay-Sachs disease and Sandhoff disease are glycolipid storage diseases caused by the lack of lysosomal β-hexosaminidase (Gravel et al., in The Metabolic Basis of Inherited Disease, eds. Scriver et al., McGraw-Hill, New York, pp. 2839- 2879, 1995). In both disorders, GM2 ganglioside and related glycolipidssubstrates for β-hexosaminidase accumulate in the nervous system and trigger acute neurodegeneration. In the most severe forms, the onset of symptoms begins in early infancy. A precipitous neurodegenerative course then ensues, with affected infants exhibiting motor dysfunction, seizure, visual loss, and deafness. Death usually occurs by 2-5 years of age. Neuronal loss through an apoptotic mechanism has been demonstrated (Huang et al., Hum. MoI. Genet. 6: 1879- 1885, 1997).
It is well-known that apoptosis plays a role in AIDS pathogenesis in the immune system. However, HIV-I also induces neurological disease. Shi et al. (J. Clin. Invest. 98: 1979- 1990, 1996) examined apoptosis induced by HlV-I infection of the CNS in an in vitro model and in brain tissue from AIDS patients, and found that HIV-I infection of primary brain cultures induced apoptosis in neurons and astrocytes in vitro. Apoptosis of neurons and astrocytes was also detected in brain tissue from 10/1 1 AIDS patients, including 5/5 patients with HIV-I dementia and 4/5 nondemented patients.
There are four main peripheral neuropathies associated with HIV, namely sensory neuropathy, AIDP/CIPD, drug-induced neuropathy and CMV-related.
The most common type of neuropathy associated with AIDS is distal symmetrical polyneuropathy (DSPN). This syndrome is a result of nerve degeneration and is characterized by numbness and a sensation of pins and needles. DSPN causes few serious abnormalities and mostly results in numbness or tingling of the feet and slowed reflexes at the ankles. It generally occurs with more severe immunosuppression and is steadily progressive. Treatment with tricyclic antidepressants relieves symptoms but does not affect the underlying nerve damage.
A less frequent, but more severe type of neuropathy is known as acute or chronic inflammatory demyelinating polyneuropathy (AIDP/CIDP). In AIDP/CIDP there is damage to the fatty membrane covering the nerve impulses. This kind of neuropathy involves inflammation and resembles the muscle deterioration often identified with long-term use of AZT. It can be the first manifestation of HIV infection, where the patient may not complain of pain, but fails to respond to standard reflex tests. This kind of neuropathy may be associated with seroconversion, in which case it can sometimes resolve spontaneously. It can serve as a sign of HIV infection and indicate that it might be time to consider antiviral therapy. AIDP/CIDP may be auto-immune in origin.
Drug-induced, or toxic, neuropathies can be very painful. Antiviral drugs commonly cause peripheral neuropathy, as do other drugs e.g. vincristine, dilantin (an anti-seizure medication), high-dose vitamins, isoniazid, and folic acid antagonists. Peripheral neuropathy is often used in clinical trials for antivirals as a dose-limiting side effect, which means that more drugs should not be administered. Additionally, the use of such drugs can exacerbate otherwise minor neuropathies. Usually, these drug-induced neuropathies are reversible with the discontinuation of the drug.
CMV causes several neurological syndromes in AIDS, including encephalitis, myelitis, and polyradiculopathy. Neuronal loss is also a salient feature of prion diseases, such as Creutzfeldt- Jakob disease in human, BSE in cattle (mad cow disease), Scrapie Disease in sheep and goats, and feline spongiform encephalopathy (FSE) in cats. Formulations of the invention may be useful for treating or preventing neuronal loss due to these prior diseases.
In another embodiment, a formulation of the invention may be used to treat or prevent any disease or disorder involving axonopathy. Distal axonopathy is a type of peripheral neuropathy that results from some metabolic or toxic derangement of peripheral nervous system (PNS) neurons. It is the most common response of nerves to metabolic or toxic disturbances, and as such may be caused by metabolic diseases • such as diabetes, renal failure, deficiency syndromes such as malnutrition and alcoholism, or the effects of toxins or drugs. The most common cause of distal axonopathy is diabetes, and the most common distal axonopathy is diabetic neuropathy. The most distal portions of axons are usually the first to degenerate, and axonal atrophy advances slowly towards the nerve's cell body. If the noxious stimulus is removed, regeneration is possible, though prognosis decreases depending on the duration and severity of the stimulus. Those with distal axonopathies usually present with symmetrical glove-stocking sensori-motor disturbances. Deep tendon reflexes and autonomic nervous system (ANS) functions are also lost or diminished in affected areas.
Diabetic neuropathies are neuropathic disorders that are associated with diabetes mellitus. These conditions usually result from diabetic microvascular injury involving small blood vessels that supply nerves (vasa nervorum). Relatively common conditions which may be associated with diabetic neuropathy include third nerve palsy; mononeuropathy; mononeuritis multiplex; diabetic amyotrophy; a painful polyneuropathy; autonomic neuropathy; and thoracoabdominal neuropathy. Clinical manifestations of diabetic neuropathy include, for example, sensorimotor polyneuropathy such as numbness, sensory loss, dysesthesia and nighttime pain; autonomic neuropathy such as delayed gastric emptying or gastroparesis; and cranial neuropathy such as oculomotor (3rd) neuropathies or Mononeuropathies of the thoracic or lumbar spinal nerves. Peripheral neuropathy is the medical term for damage to nerves of the peripheral nervous system, which may be caused either by diseases of the nerve or from the side-effects of systemic illness. Peripheral neuropathies vary in their presentation and origin, and may affect the nerve or the neuromuscular junction. Major causes of peripheral neuropathy include seizures, nutritional deficiencies, and HIV, though diabetes is the most likely cause. Mechanical pressure from staying in one position for too long, a tumor, intraneural hemorrhage, exposing the body to extreme conditions such as radiation, cold temperatures, or toxic substances can also cause peripheral neuropathy. In an exemplary embodiment, a formulation of the invention may be used to treat or prevent multiple sclerosis (MS), including relapsing MS and monosymptomatic MS, and other demyelinating conditions, such as, for example, chromic inflammatory demyelinating polyneuropathy (CIDP), or symptoms associated therewith. MS is a chronic, often disabling disease of the central nervous system.
Various and converging lines of evidence point to the possibility that the disease is caused by a- disturbance in the immune function, although the cause of this disturbance has not been established. This disturbance permits cells of the immune system to "attack" myelin, the fat containing insulating sheath that surrounds the nerve axons located in the central nervous system ("CNS"). When myelin is damaged, electrical pulses cannot travel quickly or normally along nerve fiber pathways in the brain and spinal cord. This results in disruption of normal electrical conductivity within the axons, fatigue and disturbances of vision, strength, coordination, balance, sensation, and bladder and bowel function. As such, MS is now a common and well-known neurological disorder that is characterized by episodic patches of inflammation and demyelination which can occur anywhere in the CNS. However, almost always without any involvement of the peripheral nerves associated therewith. Demyelination produces a situation analogous to that resulting from cracks or tears in an insulator surrounding an - electrical cord. That is, when the insulating sheath is disrupted, the circuit is "short circuited" and the electrical apparatus associated therewith will function intermittently or nor at all. Such loss of myelin surrounding nerve fibers results in short circuits in nerves traversing the brain and the spinal cord that thereby result in symptoms of MS. It is further found that such demyelination occurs in patches, as opposed to along the entire CNS. In addition, such demyelination may be intermittent. Therefore, such plaques are disseminated in both time and space. It is believed that the pathogenesis involves a local disruption of the blood brain barrier which causes a localized immune and inflammatory response, with consequent damage to myelin and hence to neurons.
Clinically, MS exists in both sexes and can occur at any age. However, its most common presentation is in the relatively young adult, often with a single focal lesion such as a damage of the optic nerve, an area of anesthesia (loss of sensation), or paraesthesia (localize loss of feeling), or muscular weakness. In addition, vertigo, double vision, localized pain, incontinence, and pain in the amis and legs may occur upon flexing of the neck, as well as a large variety of less common symptoms.
An initial attack of MS is often transient, and it may be weeks, months, or years before a further attack occurs. Some individuals may enjoy a stable, relatively event free condition for a great number of years, while other less fortunate ones may experience a continual downhill course ending in complete paralysis. There is, most commonly, a series of remission and relapses, in which each relapse leaves a patient somewhat worse than before. Relapses may be triggered by stressful events, viral infections or toxins. Therein, elevated body temperature, i.e., a fever, will make the condition worse, or as a reduction of temperature by, for example, a cold bath, may make the condition better.
In yet another embodiment, a formulation of the invention may be used to treat trauma to the nerves, including, trauma due to disease, injury (including surgical intervention), or environmental trauma (e.g., neurotoxins, alcoholism, etc.).
Formulations of the invention may also be useful to prevent, treat, and alleviate symptoms of various PNS disorders, such as the ones described below. The PNS is composed of the nerves that lead to or branch off from the spinal cord and CNS. The peripheral nerves handle a diverse array of functions in the body, including sensory, motor, and autonomic functions. When an individual has a peripheral neuropathy, nerves of the PNS have been damaged. Nerve damage can arise from a number of causes, such as disease, physical injury, poisoning, or malnutrition. These agents may affect either afferent or efferent nerves. Depending on the cause of damage, the nerve cell axon, its protective myelin sheath, or both may be injured or destroyed.
The term "peripheral neuropathy" encompasses a wide range of disorders in which the nerves outside of the brain and spinal cord — peripheral nerves — have been damaged. Peripheral neuropathy may also be referred to as peripheral neuritis, or if many nerves are involved, the terms polyneuropathy or polyneuritis may be used.
Peripheral neuropathy is a widespread disorder, and there are many underlying causes. Some of these causes are common, such as diabetes, and others are extremely rare, such as acrylamide poisoning and certain inherited disorders. The most common worldwide cause of peripheral neuropathy is leprosy. Leprosy is caused by the bacterium Mycobacterium leprae, which attacks the peripheral nerves of affected people. Leprosy is extremely rare in the United States, where diabetes is the most commonly known cause of peripheral neuropathy. It has been estimated that more than 17 million people in the United States and Europe have diabetes-related polyneuropathy. Many neuropathies are idiopathic; no known cause can be found. The most common of the inherited peripheral neuropathies in the United States is Charcot-Marie-Tooth disease, which affects approximately 125,000 persons. Another of the better known peripheral neuropathies is Guillain-Barre syndrome, which arises from complications associated with viral illnesses, such as cytomegalovirus, Epstein-Barr virus, and human immunodeficiency virus (HIV), or bacterial infection, including Campylobacter jejuni and Lyme disease. The worldwide incidence rate is approximately 1.7 cases per 100,000 people annually. Other well-known causes of peripheral neuropathies include chronic alcoholism, infection of the varicella-zoster virus, botulism, and poliomyelitis. Peripheral neuropathy may develop as a primary symptom, or it may be due to another disease. For example, peripheral neuropathy is only one symptom of diseases such as amyloid neuropathy, certain cancers, or inherited neurologic disorders. Such diseases may affect the PNS and the CNS, as well as other body tissues. Other PNS diseases treatable with formulations of the invention include: Brachial Plexus Neuropathies (diseases of the cervical and first thoracic roots, nerve trunks, cords, and peripheral nerve components of the brachial plexus. Clinical manifestations include regional pain, paresthesia; muscle weakness, and decreased sensation in the upper extremity. These disorders may be associated with trauma, including birth injuries; thoracic outlet syndrome; neoplasms, neuritis, radiotherapy; and other conditions. See Adams et al., Principles of Neurology, 6th ed, ppl351-2); Diabetic Neuropathies (peripheral, autonomic, and cranial nerve disorders that are associated with diabetes mellitus). These conditions usually result from diabetic microvascular injury involving small blood vessels that supply nerves (vasa nervorum). Relatively common conditions which may be associated with diabetic neuropathy include third nerve palsy; mononeuropathy; mononeuritis multiplex; diabetic amyotrophy; a painful polyneuropathy-; autonomic neuropathy; and thoracoabdominal neuropathy (see Adams et al., Principles of Neurology, 6th ed, pi 325); mononeuropathies (disease or trauma involving a single peripheral nerve in isolation, or out of proportion to evidence of diffuse peripheral nerve dysfunction). Mononeuritis multiplex refers to a condition characterized by multiple isolated nerve injuries. Mononeuropathies may result from a wide variety of causes, including ischemia; traumatic injury; compression; connective tissue diseases; cumulative trauma disorders; and other conditions; Neuralgia (intense or aching pain that occurs along the course or distribution of a peripheral or cranial nerve); Peripheral Nervous System Neoplasms (neoplasms which arise from peripheral nerve tissue). This includes neurofibromas; Schwannomas; granular cell tumors; and malignant peripheral nerve sheath tumors (see DeVita Jr et al., Cancer: Principles and Practice of Oncology, 5th ed, ppl 750-1 ); and Nerve Compression Syndromes (mechanical compression of nerves or nerve roots from internal or external causes). These may result in a conduction block to nerve impulses, due to, for example, myelin sheath dysfunction, or axonal loss. The nerve and nerve sheath injuries may be caused by ischemia; inflammation; or a direct mechanical effect; Neuritis (a general term indicating inflammation of a peripheral or cranial nerve). Clinical manifestation may include pain; paresthesias; paresis; or hyperesthesia; Polyneuropathies (diseases of multiple peripheral nerves). The various forms are categorized by the type of nerve affected (e.g., sensory, motor, or autonomic), by the distribution of nerve injury (e.g., distal vs. proximal), by nerve component primarily affected (e.g., demyelinating vs. axonal), by etiology, or by pattern of inheritance.
In another embodiment, a formulation of the invention may be used to treat or prevent chemotherapeutic induced neuropathy. The formulation of the invention may be administered prior to administration of the chemotherapeutic agent, concurrently with administration of the chemotherapeutic drug, and/or after initiation of administration of the chemotherapeutic drug. If the formulation of the invention is administered after the initiation of administration of the chemotherapeutic drug, it is desirable that the formulation of the invention be administered prior to, or at the first signs, of chemotherapeutic induced neuropathy.
Chemotherapy drugs can damage any part of the nervous system. Encephalopathy and myelopathy are fortunately very rare. Damage to peripheral nerves is much more common and can be a side effect of treatment experienced by people with cancers, such as lymphoma. Most of the neuropathy affects sensory rather than motor nerves. Thus, the common symptoms are tingling, numbness or a loss of balance. The longest nerves in the body seem to be most sensitive hence the fact that most patients will report numbness or pins and needles in their hands and feet. The chemotherapy drugs which are most commonly associated with neuropathy, are the Vinca alkaloids (anti-cancer drugs originally derived from a member of the periwinkle - the Vinca plant genus) and a platinum- containing drug called Cisplatin. The Vinca alkaloids include the drugs vinblastine, vincristine and vindesine. Many combination chemotherapy treatments for lymphoma for example CHOP and CVP contain vincristine, which is the drug known to cause this problem most frequently. Indeed, it is the risk of neuropathy that limits the dose of vincristine that can be administered.
Studies that have been performed have shown that most patients will lose some reflexes in their legs as a result of treatment with vincristine and many will experience some degree of tingling (paresthesia) in their fingers and toes. The neuropathy does not usually manifest itself right at the start of the treatment but generally comes on over a period of a few weeks. It is not essential to stop the drug at the first onset of symptoms, but if the neuropathy progresses this may be necessary. It is very important that patients should report such symptoms to their doctors, as the nerve damage is largely reversible if the drug is discontinued. Most doctors will often reduce the dose of vincristine or switch to another form of Vinca alkaloid such as vinblastine or vindesine if the symptoms are mild. Occasionally, the nerves supplying the bowel are affected causing abdominal pain and constipation. In another embodiment, a formulation of the invention may be used to treat or prevent a polyglutamine disease. Huntington's Disease (HD) and Spinocerebellar ataxia type 1 (SCAl) are just two examples of a class of genetic diseases caused by dynamic mutations involving the expansion of triplet sequence repeats. In reference to this common mechanism, these disorders are called trinucleotide repeat diseases. At least 14 such diseases are known to affect human beings. Nine of them, including SCAl and Huntington's disease, have CAG as the repeated sequence (see Table 2 below). Since CAG codes for an amino acid called glutamine, these nine trinucleotide repeat disorders are collectively known as polyglutamine diseases.
Although the genes involved in different polyglutamine diseases have little in common, the disorders they cause follow a strikingly similar course. Each disease is characterized by a progressive degeneration of a distinct group of nerve cells. The major symptoms of these diseases are similar, although not identical, and usually affect people in midlife. Given the similarities in symptoms, the polyglutamine diseases are hypothesized to progress via common cellular mechanisms. In recent years, scientists have made great strides in unraveling what the mechanisms are.
Above a certain threshold, the greater the number of glutamine repeats in a protein, the earlier the onset of disease and the more severe the symptoms. This suggests that abnormally long glutamine tracts render their host protein toxic to nerve cells.
To test this hypothesis, scientists have generated genetically engineered mice expressing proteins with long polyglutamine tracts. Regardless of whether the mice express full-length proteins or only those portions of the proteins containing the polyglutamine tracts, they develop symptoms of polyglutamine diseases. This suggests that a long polyglutamine tract by itself is damaging to cells and does not have to be part of a functional protein to cause its damage. For example, it is thought that the symptoms of SCAl are not directly caused by the loss of normal ataxin-1 function but rather by the interaction between ataxin- 1 and another protein called LANP. LANP is needed for nerve cells to communicate with one another and thus for their survival. When the mutant ataxin-1 protein accumulates inside nerve cells, it "traps'- the LANP protein, interfering with its normal function. After a while, the absence of LANP function appears to cause nerve cells to malfunction.
Table 2. Summary of Polyglutamine Diseases.
Disease Gene Chromosomal Pattern of Protein Normal Disease name location inheritance repeat repeat length length
Spinobulbar AR Xq 13-21 X-linked androgen 9-36 38-62 muscular recessive receptor atrophy (AR)
(Kennedy disease)
Huntington's HD 4pl 6.3 autosomal huntingtin 6-35 36-121 disease dominant
Dentatorubral- DRPLA 12pl 3.31 autosomal atrophin- 6-35 49-88 pallidoluysian dominant 1 atrophy (Haw
River syndrome)
Spinocerebellar SCAl 6p23 autosomal ataxin-1 6-44 39-82 ataxia type 1 dominant
Spinocerebellar SCA2 12q24.1 autosomal ataxin-2 15-31 36-63 ataxia type 2 dominant
Spinocerebellar SCA 3 14q32.1 autosomal ataxin-3 12-40 55-84 ataxia type 3 dominant
(Machado-
Joseph disease)
Spinocerebellar SCA 6 19pl3 autosomal α l A- 4-18 21-33 ataxia type 6 dominant voltage- dependent calcium channel subunit
Spinocerebellar SCA 7 3pl 2-13 autosomal ataxin-7 4-35 37-306 ataxia type 7 dominant
Spinocerebellar SCA 17 6q27 autosomal TATA 25-42 45-63 ataxia type 17 dominant binding protein Many transcription factors have also been found in neuronal inclusions in different diseases. It is possible that these transcription factors interact with the polyglutamine-containing proteins and then become trapped in the neuronal inclusions. This in turn might keep the transcription factors from turning genes on and off as needed by the cell. Another observation is hypoacetylation of histones in affected cells. This has led to the hypothesis that Class I/II Histone Deacetylase (HDAC I/II) inhibitors, which are known to increase histone acetylation, may be a novel therapy for polyglutamine diseases (US Patent Publication No. 2004/0142859; "Method of treating neurodegenerative, psychiatric, and other disorders with deacetylase inhibitors").
In yet another embodiment, the invention provides a method for treating or preventing neuropathy related to ischemic injuries or diseases, such as, for example, coronary heart disease (including congestive heart failure and myocardial infarctions), stroke, emphysema, hemorrhagic shock, peripheral vascular disease (upper and lower extremities) and transplant related injuries.
In certain embodiments, the invention provides a method to treat a central nervous system cell to prevent damage in response to a decrease in blood flow to the cell. Typically the severity of damage that may be prevented will depend in large part on the degree of reduction in blood flow to the cell and the duration of the reduction. By way of example, the normal amount of perfusion to brain gray matter in humans is about 60 to 70 mL/100 g of brain tissue/min. Death of central nervous system cells typically occurs when the flow of blood falls below approximately 8-10 mL/100 g of brain tissue/min, while at slightly higher levels (i.e. 20-35 mL/100 g of brain tissue/min) the tissue remains alive but not able to function. In one embodiment, apoptotic or necrotic cell death may be prevented. In still a further embodiment, ischemic-mediated damage, such as cytoxic edema or central nervous system tissue anoxemia, may be prevented. In each embodiment, the central nervous system cell may be a spinal cell or a brain cell.
Another aspect encompasses administrating a formulation of the invention to a subject to treat a central nervous system ischemic condition. A number of central nervous system ischemic conditions may be treated by the formulations of the invention. In one embodiment, the ischemic condition is a stroke that results in any type of ischemic central nervous system damage, such as apoptotic or necrotic cell death, cytoxic edema or central nervous system tissue anoxia. The stroke may impact any area of the brain or be caused by any etiology commonly known to result in the occurrence of a stroke. In one alternative of this embodiment, the stroke is a brain stem stroke. Generally speaking, brain stem strokes strike the brain stem, which control involuntary life-support functions such as breathing, blood pressure, and heartbeat. In another alternative of this embodiment, the stroke is a cerebellar stroke. Typically, cerebellar strokes impact the cerebellum area of the brain, which controls balance and coordination. In still another embodiment, the stroke is an embolic stroke. In general terms, embolic strokes may impact any region of the brain and typically result from the blockage of an artery by a vaso-occlusion. In yet another alternative, the stroke may be a hemorrhagic stroke. Like ischemic strokes, hemorrhagic stroke may impact any region of the brain, and typically result from a ruptured blood vessel characterized by a hemorrhage (bleeding) within or surrounding the brain. In a further embodiment, the stroke is a thrombotic stroke. Typically, thrombotic strokes result from the blockage of a blood vessel by accumulated deposits.
In another embodiment, the ischemic condition may result from a disorder that occurs in a part of the subject's body outside of the central nervous system, but yet still causes a reduction in blood flow to the central nervous system. These disorders may include, but are not limited to a peripheral vascular disorder, a venous thrombosis, a pulmonary embolus, arrhythmia (e.g. atrial fibrillation), a myocardial infarction, a transient ischemic attack, unstable angina, or sickle cell anemia. Moreover, the central nervous system ischemic condition may occur as result of the subject undergoing a surgical procedure. By way of example, the subject may be undergoing heart surgery, lung surgery, spinal surgery; brain surgery, vascular surgery, abdominal surgery, or organ transplantation surgery. The organ transplantation surgery may include heart, lung, pancreas, kidney or liver transplantation surgery. Moreover, the central nervous system ischemic condition may occur as a result of a trauma or injury to a part of the subject's body outside the central nervous system. By way of example, the trauma or injury may cause a degree of bleeding that significantly reduces the total volume of blood in the subject's body. Because of this reduced total volume, the amount of blood flow to the central nervous system is concomitantly reduced. By way of further example, the trauma or injury may also result in the formation of a vaso-occlusion that restricts blood flow to the central nervous system. Of course it is contemplated that the formulations of the invention may be employed to treat the central nervous system ischemic condition irrespective of the cause of the condition. In one embodiment, the ischemic condition results from a vaso-occlusion. The vaso-occlusion may be any type of occlusion, but is typically a cerebral thrombosis or an embolism. In a further embodiment, the ischemic condition may result from a hemorrhage. The hemorrhage may be any type of hemorrhage, but is generally a cerebral hemorrhage or a subararachnoid hemorrhage. In still another embodiment, the ischemic condition may result from the narrowing of a vessel. Generally speaking, the vessel may narrow as a result of a vasoconstriction such as occurs during vasospasms, or due to arteriosclerosis. In yet another embodiment, the ischemic condition results from an injury to the brain or spinal cord.
In yet another aspect, a formulation of the invention may be administered to reduce infarct size of the ischemic core following a central nervous system ischemic condition. Moreover, a formulation of the invention may also be beneficially administered to reduce the size of the ischemic penumbra or transitional zone following a central nervous system ischemic condition.
In one embodiment, a combination drug regimen may include drugs or compounds for the treatment or prevention of neurodegenerative disorders or secondary conditions associated with these conditions. Thus, a combination drug regimen may include a formulation of the invention and one or more anti- neurodegeneration agents. For example, a formulation of the invention can be combined with an effective amount of one or more of: L-DOPA; a dopamine agonist; an adenosine A2A receptor antagonist; a COMT inhibitor; a MAO inhibitor; an N-NOS inhibitor; a sodium channel antagonist; a selective N -methyl D- aspartate (NMDA) receptor antagonist; an AMPA/kainate receptor antagonist; a calcium channel antagonist; a GABA-A receptor agonist; an acetyl-choline esterase inhibitor; a matrix metalloprotease inhibitor; a PARP inhibitor; an inhibitor of p38 MAP kinase or c-jun-N-terminal kinases; TPA; NDA antagonists; beta-interferons; growth factors; glutamate inhibitors; and/or as part of a cell therapy.
Exemplary N-NOS inhibitors include 4-(6-amino-pyridin-2-yl)-3- methoxyphenol 6-[4-(2-dimethylamino-ethoxy)-2-methoxy-phenyl]-pyridin-2-yl- amine, 6-[4-(2-dimethylamino-ethoxy)-2,3-dimet-hyl-phenyl]-pyridin-2-yl-amine, 6-[4-(2-pyrrolidinyl-ethoxy)-2,3-dimethyl-p-henyl]-pyridin-2-yl-amine, 6-[4-(4-(n- methyl)piperidinyloxy)-2,3-dimethyl-p-henyl]-pyridin-2-yl-amine, 6-[4-(2- dimethylamino-ethoxy)-3-methoxy-phenyl]-pyridin-2-yl-amine, 6-[4-(2- pyrrolidinyl-ethoxy)-3-methoxy-phenyl]-pyridin-2-yl-amine, 6-{4-[2-(6,7- dimethoxy-3,4-dihydro-l h-isoquinolin-2-yl)-ethoxy]-3-methoxy-phenyl}-pyridin-2- yl-amine, 6-{3-methoxy-4-[2-(4-phenethyl-piper-azin-l-yl)-ethoxy]-phenyl}- pyridin-2-yl-amine, 6-{3-methoxy-4-[2-(4-methyl-piperazin-l-yl)-ethoxy]-phenyl}- pyridin-2-yl-amine, 6-{4-[2-(4-dimethylarnin-o-piperidin-l -yl)-ethoxy]-3-methoxy- phenyl}-pyridin-2-yl-amine, 6-[4-(2-dimethylamino-ethoxy)-3-ethoxy-phenyl]- pyridin-2-yl-amine, 6-[4-(2-pyiτolidinyl-ethoxy)-3-ethoxy-phenyl]-pyridin-2-yl- amine, 6-[4-(2-dimethylamino-ethoxy)-2-isopiOpyl-phenyl]-pyridin-2-yl-amine, 4- (6-amino-pyridin-yl)-3-cyclopropyl-phenol 6-[2-cyclopropyl-4-(2-dimethy-lamino- ethoxy)-phenyl]-pyridin-2-yl-amine, 6-[2-cyclopropyl-4-(2-pyrrolidin- 1 -yl-ethoxy)- phenyl]-pyridin-2-yl-amine, 3-[3-(6-amino-pyridin-2yl)-4-cycl-opropyl-phenoxy]- pyrrolidine- 1 -carboxylic acid tert-butyl ester 6-[2-cyclopropyl-4-(l -methyl- pyrrolidin-3-yl-oxy)-phenyl]-pyridin-2-yl-amine, 4-(6-amino-pyridin-2-yl)-3- cyclobutyl-phenol 6-[2-cyclobutyl-4-(2-dime-thylamino-ethoxy)-phenyl]-pyridin-2- yl-amine. 6-[2-cyclobutyl-4-(2-pyrrolid-in- 1 -yl-ethoxy)-phenyl]-pyridin-2-yl-amine, 6-[2-cyclobutyl-4-(l -methyl-pyr-rolidin-3-yl-oxy)-phenyl] -pyridin-2-yl-amine, 4-(6- amino-pyridin-2-yl)-3-cy-clopentyl-phenol 6-[2-cyclopentyl-4-(2-dimethylamino- ethoxy)-phenyl]-pyrid-in-2-yl-amine, 6-[2-cyclopentyl-4-(2-pyrrolidin- 1 yl-ethoxy)- phenyl]-pyridin-2-yI-amine, 3-[4-(6-amino-pyridin-2yl)-3-methoxy-phenoxy]- pyrrolidine-1 -ca-rboxylic acid tert butyl ester 6-[4-( l -methyl-pyrrolidin-3-yl-oxy)-2- metho-xy-phenyl]-pyridin-2-yl-amine, 4-[4-(6-amino-pyridin-2yl)-3-methoxy- phenoxy-]-piperidine- 1 -carboxylic acid tert butyl ester 6-[2-methoxy-4-( 1 -methyl-p- iperidin-4-yl-oxy)-phenyl]-pyridin-2-yl-amine, 6-[4-(allyloxy)-2-methoxy-ph-enyl]- pyridin-2-yl-amine, 4-(6-amino-pyridin-2-yl)-3-methoxy-6-allyl-phenol 12 and 4-(6- amino-pyridin-2-yl)-3-methoxy-2-allyl-phenol 13 4-(6-amino-pyridin-2-yl)-3- methoxy-6-propyl-phenol 6-[4-(2-dimethylamino-ethoxy)-2-methoxy-5-propyl- phenyl]-pyridin-yl-amine, 6-[2-isopropyl-4-(pyrrolidin-3-yl-oxy)-phenyl]-pyridin-2- yl-amine, 6-[2-isopropyl-4-(piperidin-3-yl-oxy)-phenyl]-pyridin-2-yl-amine, 6-[2- isopropyl-4-(l -methyl-azetidin-3-yl-oxy)-phenyl]-pyridin-2-yl-amine, 6-[2- isopropyl-4-(l -methyl-piperidin-4-yl-oxy)-phenyl]-pyridin-2-yl-amine, 6-[2- isopropyl-4-(l-methyl-pyrrolidin-3-yl-oxy)-phenyl]-pyridin-2-yl-amin-e 6-[2- isopropyl-4-( 1 -methyl-pyrrolidin-3-yl-oxy)-phenyl]-pyridin-2-yl-amine, 6-[2- isopropyl-4-(2-methyl-2-aza-bicyclo[2.2.1 ]hept-5-yl-oxy)-phenyl]-p-yridin-2-yl- amine, 6-[4-(2-dimethylamino-ethoxy)-2-methoxy-phenyl]-pyridin-2-yl-amine, 6- {4-[2-(benzyl-methyl-amino)-ethoxy]-2-methoxy-phenyl}-pyridin-2-yl-amine, 6-[2- methoxy-4-(2-pyrrolidin- 1 -yl-ethoxy)-phenyl]-pyridin-2-yl-amine, 2-(6-amino- pyridin-2-yl)-5-(2-dimethylamino-ethoxy)-phenol 2-[4-(6-amino-pyridin-2-yl)-3- methoxy-phenoxy]-acetamide 6-[4-(2-amino-ethoxy)-2-methoxy-phenyl]-pyridin-2- yl-amine, 6-{4-[2-(3,4-dihydiO- l h-isoquinolin-2-yl)-ethoxy]-2-methoxy-phenyl}- pyrid-in-2-yl-amine, 2-[4-(6-amino-pyridin-2-yl)-3-methoxy-phenoxy]-ethanol 6- {2-methoxy-4-[2-(2,2,6,6-tetramethyl-piperidin- 1 -yl)-ethoxy]-phenyl] -py-ridin-2- yl-amine, 6-{4-[2-(2,5-dimethyl-pyπOlidin-l -yl)-ethoxy]-2-methoxy-phenyl}- pyridin-2-yl-amine, 6- {4-[2-(2,5-dimethyl-pyiτolidin- 1 -yl)-ethoxy]-2-methoxy- phenyl}-pyridin-2-yl-amine, 2-[4-(6-amino-pyridin-2-yl)-3-methoxy-phenoxy]-l - (2,2,6,6-tetramethyl-piperidin- 1 -yl)-ethanone 6-[2-mcthoxy-4-( 1 -methyl-pyrrolidin- 2-yl-methoxy)-phenyl]-pyridin-2-yl-amine, 6-[4-(2-dimethylamino-ethoxy)-2- propoxy-phenyl]-pyridin-2-yl-amine, 6-{4-[2-(benzyl-methyl-amino)-ethoxy]-2- piOpoxy-phenyl}-pyridin-2-yl-amin-e 6-[4-(2-ethoxy-ethoxy)-2-methoxy-phenyl]- pyridin-2-yl -amine, 6-[4-(2-dimethylamino-ethoxy)-2-isopropoxy-phcnyl]-pyridin- 2-yl-amine, 6-[4-(2-ethoxy-ethoxy)-2-isopi"opoxy-phenyl]-pyridin-2-yl-amine, 6-[2- methoxy-4-(3-methyl-butoxy)-phenyl]-pyridin-2-yl-amine, 6-[4-(2-dimethylamino- ethoxy)-2-ethoxy-phenyl]-pyridin-2-yl-amine, 6- [4-[2-(benzyl-methyl-amino)- ethoxy]-2-ethoxy-phenyl}-pyridin-2-yl-amine, 6-[2-ethoxy-4-(3-methyl-butoxy)- phenyl]-pyridin-2-yl-amine, l -(6-amino-3-aza-bicyclo[3.1.0]hex-3-yl)-2-[4-(6- amino-pyridin-2-yl)-3-et-hoxy-phenoxy]-ethanone 6-[2-ethoxy-4-(2-pyπOlidin-l -yl- ethoxy)-phenyl]-py-ridin-2-yl-amine. 3-{2-[4-(6-amino-pyridin-2-yl)-3-ethoxy- phenoxy]-ethyl}-3-aza-bicyclo[3.1.0]hex-6-yl-amine, l-(6-amino-3-aza- bicyclo[3.1.0]hex-3-yl)-2-[4-(6-amino-pyridin-2-yl)-3-methoxy-phenoxy]-ethanone 3-{2-[4-(6-amino-pyridin-2-yl)-3-methoxy-phenoxy]-ethyl}-3-aza-bicyclo[3.- 1.0]hex-6-yl-amine, 6-[2-isopropoxy-4-(2-pyrrolidin-l-yl-ethoxy)-phenyl]-py-ridin- 2-yl-amine, 6-{4-[2-(benzyl-methyl-amino)-ethoxy]-2-isopropoxy-phenyl-}-pyridin- 2-yl-amine, 6-[4-(2-dimethylamino-ethoxy)-2-methoxy-5-propyl-phen-yl]-pyridin- 2-yl-amine, 6-[5-allyl-4-(2-dimethylamino-ethoxy)-2-methoxy-phe-nyl]-pyridin-2- yl-amine, 6-[5-allyl-2-methoxy-4-(2-pyiτolidin-l-yl-ethoxy)-phenyl]-pyridin-2-yl- amine, 6-[3-allyl-4-(2-dimethylamino-ethoxy)-2-methoxy-phenyl]-pyridin-2-yl- amine, 6-[2-methoxy-4-(pyrrolidin-3-yl-oxy)-phenyl]-p-yridin-2-yl-amine, 6-[2- methoxy-4-( 1 -methyl-pyrrolidin-3-yl-oxy)-phenyl]-py-ridin-2-yl-amine, 6-[2- ethoxy-4-(pyrrolidin-3-yl-oxy)-phenyl]-pyridin-2-yl-amine, 6-[2-isopropoxy-4- (pyrrolidin-3-yl-oxy)-phenyl]-pyridin-2-yl-amine, 6-[2-methoxy-4-(piperidin-4-yl- oxy)-phenyl]-pyridin-2-yl-amine, 6-[2-methoxy-4-(2,2,6,6-tetramethyl-piperidin-4- yl-oxy)-phenyl]-pyridin-2-yl-amine, 6-[2-isopropoxy-4-(pyrroLidin-3-yl-oxy)- phenyl]-pyridin-2-yl-amine, 3-[4-(6-amino-pyridin-2-yl)-3-methoxy-phenoxy]- azetidine-1 -carboxylic acid tert-butyl ester 6-[4-(azetidin-3-yl-oxy)-2-methoxy- phenyl]-pyridin-2-yl-amine, 6-[2-methoxy-4-( 1 -methyl-azetidin-3-yl-oxy)-phenyl]- pyridin-2-y-l-amine, 6-[2-isopropoxy-4-(pyiτolidin-3-yl-oxy)-phenyl]-pyridin-2-yl- amine, 6-[2-isopropoxy-4-(pyiτolidin-3-yl-oxy)-phenyl]-pyridin-2-yl-amine, 6-[2- methoxy-4-(pyriOlidin-3-yl-oxy)-phenyl]-pyridin-2-yl-amine, 6-[2-methoxy-4-( 1 - methyl-pyrrolidin-3-yl-oxy)-phenyl]-pyridin-2-yl-amine, 6-[2-methoxy-4-( l -methyl- pyiτolidin-3-yl-oxy)-phenyl]-pyridin-2-yl-amine, 6-[2-methoxy-4-(2-methyl-2-aza- bicyclo[2.2.1 ]hept-5-yl-oxy)-phenyl]-pyrid-in-2-yl-amine, 6-[2-methoxy-4-(l - methyl-piperidin-4-yl-oxy)-phenyl]-pyridin-2-yl-amine, 6-[4-(l -ethyl-piperidin-4- yl-oxy)-2-methoxy-phenyl]-pyridin-2-yl-amine, 6-[5-allyl-2-methoxy-4-( l -methyl- pyriOlidin-3-yl-oxy)-phenyl]-pyr-idin-2-yI-amine, 6-[4-(2-dimethylamino-ethoxy)- 2,6-dimethyl-phenyl]-pyridin-2-yl-amine, 6-[2,6-dimethyl-4-(3-piperidin- 1 -yl- propoxy)-phenyl]-pyridin-2-yl-amine, 6-[2,6-dimethyl-4-(2-pyiTolidin- l -yl-ethoxy)- phenyl]-pyridin-2-y-l-amine, 6-{2,6-dimethyl-4-[3-(4-methyl-piperazin-l -yl)- propoxy]-phenyl}-py-ridin-2-yl-amine, 6-[2,6-dimethyl-4-(2-morpholin-4-yl- ethoxy)-phenyl]-pyrid-in-2-yl-amine, 6-{4-[2-(benzyl-methyl-amino)-ethoxy]-2,6- dimethyl-phenyl}-p-yridin-2-yl-amine, 2-[4-(6-amino-pyridin-2-yl)-3,5-dimethyl- phenoxy]-acetam-ide 6-[4-(2-amino-ethoxy)-2,6-dimethyl-phenyl]-pyridin-2-yl- amine, 6-[2-isopropyl-4-(2-pyrrolidin- 1 -yl-ethoxy)-phenyl]-pyridin-2-yl-amine, 2- (2,5-dimethyl-pyrrolidin- 1 -yl)-6-[2-isopropyl-4-(2-pyrrolidin- 1 -yl-etho-xy)-phenyl]- pyridine 6- {4-[2-(3,5-dimethyl-piperidin-l-yl)-ethoxy]-2-isopr-opyl-phenyl}- pyridin-2-yl-amine, 6-[4-(2-dimethylamino-ethoxy)-2-isopropyl-phenyl]-pyridin-2- yl-amine, 6-[2-tert-butyl-4-(2-dimethylamino-ethoxy)-phen-yl]-pyridin-2-yl-amine, 6-[2-tert-butyl-4-(2-pyrrolidin-l -yl-ethoxy)-phenyl-]-pyridin-2-yl-amine, 6-[4-(2- pyrrolidinyl-ethoxy)-2,5-dimethyl-phenyl]-pyr-idin-2-yl-amine, 6-[4-(2- dimethylamino-ethoxy)-2,5-dimethyl-phenyl]-pyridin-2-yl-amine, 6-[4-(2-(4- phenethylpiperazin-l-yl)-ethoxy)-2,5-dimethyl-pheny-l]-pyridin-2-yl-amine, 6-[2- cyclopropyl-4-(2-dimethylamino- 1 -methyl-ethoxy)-phenyl]-pyridin-2-yl-amine, 6- [cyclobutyl-4-(2-dimethylamino- 1 -methyl-etho-xy)-phenyl]-pyridin-2-yl-amine, 6- [4-(allyloxy)-2-cyclobutyl-phenyl]-pyridi-n-2yl amine, 2-allyl-4-(6-amino-pyridin-2- yl)-3-cyclobutyl-phenol and 2-allyl-4-(6-amino-pyridin-2-yl)-5-cyclobutyl-phenol A- (6-amino-pyridin-2yl)-5-cyclobutyl-2-propyl-phenol 4-(6-amino-pyridin-2yl)-3- cyclobutyl-2-propyl-phenol 6-[2-cyclobutyl-4-(2-dimethylamino-l -methyl-ethoxy)- 5-propyl-phenyl]-pyri-din-2-yl-amine, 6-[2-cyclobutyl-4-(2-dimethylamino- 1 - methyl-ethoxy)-3-propy-l-phenyl]-pyridin-2-yl-amine, 6-[2-cyclobutyl-4-(2- dimethylamino-ethoxy)-5-propyl-phenyl]-pyridin-2-yl-amine, 6-[2-cyclobutyl-4-(2- dimethylamino-ethox-y)-3-propyl-phenyl]-pyridin-2-yl-amine, 6-[2-cyclobutyl-4-( 1 - methyl-pyπOli-din-3-yl-oxy)-5-piOpyl-phenyl]-pyridin-2-yl-amine, 6-[cyclobutyl-4- ( 1 -methy-l-pyrrolidin-3-yl-oxy)-3-propyl-phenyl]-pyridin-2-yl-amine, 2-(4- benzyloxy-5-hydiOxy-2-methoxy-phenyl)-6-(2,5-dimethyl-pyiτol-l -yl)-p-yridine 6- [4-(2-dimethylamino-ethoxy)-5-ethoxy-2-methoxy-phenyl]-pyridin-2-yl -amine, 6-
[5-ethyl-2-methoxy-4-( l -methyl-piperidin-4-yl-oxy)-phenyl]-pyr-idin-2-yl-amine, 6-' [5-ethyl-2-methoxy-4-(piperidin-4-yl-oxy)-phenyl]-pyridi-n-2-yl-amine, 6-[2,5- dimethoxy-4-( l -methyl-pyriOlidin-3-yl-oxy)-phenyl]-pyr-idin-2-yl-amine, 6-[4-(2- dimethylamino-ethoxy)-5-ethyl-2-methoxy-phenyl]-py-ridin-2-yl-amine. Exemplary NMDA receptor antagonists include (+)-( 1 S, 2S)- 1 -(4-hydroxy- phenyl)-2-(4-hydroxy-4-phenylpiperidino)- l -pro-panol, (I S, 2S)-l -(4-hydroxy-3- methoxyphenyl)-2-(4-hydroxy-4-phenylpiperi-dino)-l -propanol, (3R, 4S)-3-(4-(4- fluorophenyl)-4-hydroxypiperidin-l-yl-)-chroman-4,7-diol, (IR*, 2R*)-l -(4- hydroxy-3-methylphenyl)-2-(4-(4-fluoro-phenyl)-4-hydroxypiperidin-l-yl)-propan- 1 -ol-mesylate or a pharmaceutically acceptable acid addition salt thereof.
Exemplary dopamine agonists include ropininole; L-dopa decarboxylase inhibitors such as carbidopa or benserazide, bromocriptine, dihydroergocryptine, etisulergine, AF- 14, alaptide, pergolide, piribedil; dopamine Dl receptor agonists such as A-68939, A-77636, dihydrexine, and SKF-38393; dopamine D2 receptor agonists such as carbergoline, lisuride, N-0434, naxagolide, PD-1 18440, pramipexole, quinpirole and ropinirole; dopamine/β-adrenegeric receptor agonists such as DPDMS and dopexamine; dopamine/5-HT uptake inhibitor/5-HT- 1 A agonists such as roxindole; dopamine/opiate receptor agonists such as NIH- 10494; α2-adrenergic antagonist/dopamine agonists such as terguride; α2-adrenergic antagonist/dopamine D2 agonists such as ergolines and talipexole; dopamine uptake inhibitors such as GBR-12909, GBR-13069, GYKI-52895, and NS-2141 ; monoamine oxidase-B inhibitors such as selegiline, N-(2-butyl)-N- methylpropargylamine, N-methyl-N-(2-pentyl)propargylamine, AGN-1 133, ergot derivatives, lazabemide, LU-53439, MD-280040 and mofegiline; and COMT inhibitors such as CGP-28014.
Exemplary acetyl cholinesterase inhibitors include donepizil, 1 -(2-methyl- lH-benzimida-zol-5-yl)-3-[ l -(phenylmethyl)-4-piperidinyl]-l -propanone; l -(2- phenyl- 1 H-benzimidazol-5-yl)-3-[ 1 -(phenylmethyl)-4-piperidinyl]- 1 -pr-opanone; 1 - ( 1 -ethyl-2-methyl- 1 H-benzimidazol-5-yl)-3 -[ 1 -(phenylmethyl)-4-p-iperidinyl]- 1 - propanone; 1 -(2-methyl-6-benzothiazolyl)-3-[ 1 -(phenylmethyl)-4-piperidinyl]- 1 - propanone; 1 -(2-methyl-6-benzothiazolyl)-3-[ 1 -[(2-methyl-4-thiazolyl)methyl]-4- piperidinyl]- 1 -propanone; 1 -(5-methyl-benzo[b]thie-n-2-yl)-3-[ 1 -(phenylmethyl)4- piperidinyl]- 1 -propanone; 1 -(6-methyl-benzo[b]thien-2-yl)-3-[ 1 -(phenylmethyl)-4- piperidinyl]- l -prop-anone; l -(3,5-dimethyl-benzo[b]thien-2-yl)-3-[l - (phenylmethyl)-4-piperidin-yl]-l -propanone; l -(benzo[b]thien-2-yl)-3-[ l - (phenylmethyl)-4-piperidinyl]-l -propanone; l -(benzofuran-2-yl)-3-[l - (phenyImethyl)-4-piperidinyl]-l -pro-panone; 1 -( 1 -phenylsulfonyl-6-methyl-indol-2- yl)-3-[ 1 -(phenylmethyl)-4-pip-eridinyl]- 1 -propanone; 1 -(6-methyl-indol-2-yl)-3-[ 1 - (phenylmethyl)-4-piper-idinyl]-l -propanone; l -(l -phenylsulfonyl-5-amino-indol-2- yl)-3-[l-(phenylm-ethyl)-4-piperidinyl]-l-propanone; l-(5-amino-indol-2-yl)-3-[l - (phenylmet-hyl)-4-piperidinyl]-l-propanone; and l -(5-acetylamino-indol-2-yl)-3-[l- (ph-enylmethyl)-4-piperidinyl]- 1 -propanone. 1 -(6-quinolyl)-3-[ 1 -(phenylmethyl)-4- piperidinyl]-l-propanone; l -(5-indolyl)-3-[l -(phenylmethyl)-4-piperidiny-l]-l- propanone; l -(5-benzthienyl)-3-[l-(phenylmethyl)-4-piperidinyl]-l-pro-panone; 1 - (6-quinazolyl)-3-[l -(phenylmethyl)-4-piperidinyl]-l -propanone; l-(6- benzoxazolyl)-3-[l -(phenylmethyl)-4-piperidinyl]-l -propanone; l-(5-benzofuryl)-3- [l -(phenylmethyl)-4-piperidinyl]-l -propanone; l-(5-methyl-benzimidazol-2-yl)-3- [ 1 -(phenylmethyl)-4-piperidinyl]- 1 -propa-none; 1 -(6-methyl-benzimidazol-2-yl)-3- [l -(phenylmethyl)-4-piperidinyl]-l -propanone; l-(5-chloro-benzo[b]thien-2-yl)-3- [l-(phenylmethyl)-4-piperidin-yl]-l -propanone; l-(5-azaindol-2-yl)-3-[l- (phenylmethyl)4-piperidinyl]-l-p-ropanone; l -(6-azabenzo[b]thien-2-yl)-3-[l- (phenylmethyl)-4-piperidinyl]-l -propanone; l -(lH-2-oxo- pyrrolo[2',3',5,6]benzo[b]thieno-2-yl)-3-[l -(phenylmethyl)-4-piperidinyl]-l - propanone; 1 -(6-methyl-benzothiazol-2-yl)-3-[ 1 -(phenylmethyl)-4-piperidinyl]- 1 - propanone; l -(6-methoxy-indol-2-yl)-3-[l -(phenylmethyl)-4-piperidinyl]-l - propanone; 1 -(6-methoxy-benzo[b]thien-2-yl)-3-[ 1 -(phenylmethyl)-4-piperidinyl]- 1 - pro-panone; 1 -(6-acetylamino-benzo[b]thien-2-yl)-3-[ 1 -(phenylmethyl)-4-piperid- inyl]-l -propanone; l -(5-acetylamino-benzo[b]thien-2-yl)-3-[ l -(phenylmethyl-)-4- piperidinyl]- 1 -propanone; 6-hydroxy-3-[2-[ 1 -(phenylmethyl)-4-piperidin-yl]ethyl]- 1 ,2-benzisoxazole; 5-methyl-3-[2-[ l -(phenylmethyl)-4-piperidinyl-]ethyl]-l ,2- benzisoxazole; 6-methoxy-3[2-[l(phenylmethyl)-4-piperidinyl]et-hyl]- l ,2- benzisoxazole; 6-acetamide-3-[2-[ l -(phenylmethyl)-4-piperidinyl]-ethyl]-l ,2- benzisoxazole; 6-amino-3-[2-[l -(phenymethyl)-4-pipcridinyl]ethy-l]- l ,2- benzisoxazole; 6-(4-morpholinyl)-3-[2-[ 1 -(phenylmethyl)-4-piperidin-yl]ethyl]- 1 ,2- benzisoxazole; 5,7-dihydro-3-[2-[ 1 -(phenylmethyl)-4-piperidi-nyl]ethyl]-6H- pyrrolo[4,5-f]-l ,2-benzisoxazol-6-one; 3-[2-[ 1 -(phenylmethyl)-4-piperidinyl]ethyl]- 1 ,2-benzisothiazole; 3-[2-[ 1 -(phenylmethyl)-4-piperidinyl]ethenyl]- 1 ,2- benzisoxazole; 6-phenylamino-3-[2-[ 1 -(phenylmethyl)-4-piperidinyl]ethyl]- 1 ,2,- benzisoxaz-ole; 6-(2-thiazoly)-3-[2-[ 1 -(phenylmethyl)-4-piperidinyl]ethyl]- 1 ,2- benzis-oxazole; 6-(2-oxazolyl)-3-[2-[l -(phenylmethyl)-4-piperidinyl]ethyl]-l ,2-be- nzisoxazole; 6-pyrrolidinyl-3-[2-[ 1 -(phenylmethyl)-4-piperidinyl]ethyl]- 1 ,-2- benzisoxazole; 5,7-dihydro-5,5-dimethyl-3-[-2-[l-(phenylmethyl)-4-piperid- inyl]ethyl]-6H-pyrrolo[4,5-f]-l,2-benzisoxazole-6-one; 6,8-dihydro-3-[2-[l- (phenylmethyl)-4-piperidinyl]ethyl]-7H-pyrrolo[5,4-g]-l,2-benzisoxazole-7-one; 3- [2-[l-(phenylmethyl)-4-piperidinyl]ethyl]-5,6,-8-trihydro-7H-isoxazolo[4,5-g]- quinolin-7-one; l-benzyl-4-((5,6-dimethoxy-l -indanon)-2-yl)methylpiperidine, 1- benzyl-4-((5,6-dimethoxy- 1 -indanon)-2-ylidenyl)methylpiperidine, 1 -benzyl-4-((5- methoxy-l-indanon)-2-yl)methylp-iperidine, l-benzyl-4-((5,6-diethoxy-l-indanon)- 2-yl)methylpiperidine, 1 -benzyl -4-((5,6-methnylenedioxy- 1 -indanon)-2- yl)methylpiperidine, 1 -(m-nitrobenzyl)-4-((5,6-dimethoxy-l -indanon)-2- yl)methylpiperidine, 1 -cyclohexymethyl-4-((5,6-dimethoxy-l -indanon)-2- yl)methylpiperidine, l-(m-florobenzyl)-4-((5,6-dimethoxy-l-indanon)-2- yl)methylpiperidine, 1 -benzyl-4-((5,6-dimethoxy- 1 -indanon)-2-yl)propylpiperidine, and l-benzyl-4-((5-isopropoxy-6-methoxy-l-indanon)-2-yl)methylpiperidine.
Exemplary calcium channel antagonists include diltiazem, omega-conotoxin GVIA, methoxyverapamil, amlodipine, felodipine, lacidipine, and mibefradil.
Exemplary GABA-A receptor modulators include clomethiazole; IDDB; gaboxadol (4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol); ganaxolone (3α- hydroxy-3β-methyl-5α-pregnan-20-one); fengabine (2-[(butylimino)-(2- chlorophenyl)methyl]-4-chlorophenol); 2-(4-methoxyphenyl)-2,5,6,7,8,9- hexahydro-pyrazolo[4,3-c]cinnolin-3-one; 7-cyclobutyl-6-(2-methyl-2H-l ,2,4- triazol-3-ylmethoxy)-3-phenyl-l ,2,4-triazolo[4,3-b]pyridazine; (3-fluoro-4- methylphenyl)-N-({-l -[(2-methylphenyl)methyl]-benzimidazol-2-yl}methyl)-N- pentylcarboxamide; and 3-(aminomethyl)-5-methylhexanoic acid.
Exemplary potassium channel openers include diazoxide, flupirtine, pinacidil, levcromakalim, rilmakalim, chromakalim, PCO-400 and SKP-450 (2-
[2"(1 ", 3"-dioxolone)-2-methyl]-4-(2'-oxo-l '-pyrrolidinyl)-6-nitro-2H-l -benzopyra- n).
Exemplary AiMPA/kainate receptor antagonists include 6-cyano-7- nitroquinoxalin-2,3-di-one (CNQX); 6-nitro-7-sulphamoylbenzo[fjquinoxaline-2,3- dione (NBQX); 6,7-dinitroquinoxaline-2,3-dione (DNQX); 1 -(4-aminophenyl)-4- methyl-7,8-methylenedioxy-5H-2,3-benzodiazepine hydrochloride; and 2,3- dihydroxy-6-nitro-7-sulfamoylbenzo-[f]quinoxaline. Exemplary sodium channel antagonists include ajmaline, procainamide, flecainide and riluzole.
Exemplary matrix-metalloprotease inhibitors include 4-[4-(4- fluorophenoxy)benzenesulfon-ylamino]tetrahydropyran-4-carboxylic acid hydroxyamide; 5-Methyl-5-(4-(4'-fluorophenoxy)-phenoxy)-pyrimidine-2,4,6- trione; 5-n-Butyl-5-(4-(4'-fluorophenoxy)-phenoxy)-pyrimidine-2,4,6-trione and prinomistat.
PoIy(ADP ribose) polymerase (PARP) is an abundant nuclear enzyme which is activated by DNA strand single breaks to synthesize poly (ADP ribose) from NAD. Under normal conditions, PARP is involved in base excision repair caused by oxidative stress via the activation and recruitment of DNA repair enzymes in the nucleus. Thus, PARP plays a role in cell necrosis and DNA repair. PARP also participates in regulating cytokine expression that mediates inflammation. Under conditions where DNA damage is excessive (such as by acute excessive exposure to a pathological insult), PARP is over-activated, resulting in cell-based energetic failure characterized by NAD depletion and leading to ATP consumption, cellular necrosis, tissue injury, and organ damage/failure. PARP is thought to contribute to neurodegeneration by depleting nicotinamide adenine dinucleotide (NAD+) which then reduces adenosine triphosphate (ATP; Cosi and Marien, Ann. N. Y. Acad. Sci., 890:227, 1999) contributing to cell death which can be prevented by PARP inhibitors. Exemplory PARP inhibitors can be found in Southan and Szabo, Current Medicinal Chemistry, 10:321 , 2003.
Exemplary inhibitors of p38 MAP kinase and c-jun-N-teπninal kinases include pyridyl imidazoles, such as PD 169316, isomeric PD 1693 16, SB 203580, SB 202190, SB 220026, and RWJ 67657. Others are described in US Patent 6,288,089, and incorporated by reference herein.
In an exemplary embodiment, a combination therapy for treating or preventing MS comprises a formulation of the invention and one or more of Avonex® (interferon beta- I a), Tysabrr (natalizumab), or Fumaderm® (BG- 12/Or-al Fumarate).
In another embodiment, a combination therapy for treating or preventing diabetic neuropathy or conditions associated therewith comprises a therapeutically effective amount of formulation of the invention and one or more of tricyclic antidepressants (TCAs) (including, for example, imipramine, amytriptyline, desipramine and nortriptyline), serotonin reuptake inhibitors (SSRIs) (including, for example, fluoxetine, paroxetine, sertralene, and citalopram) and antiepileptic drugs (AEDs) (including, for example, gabapentin, carbamazepine, and topimirate).
In another embodiment, the invention provides a method for treating or preventing a polyglutamine disease using a combination comprising formulation of the invention and at least one HDAC I/II inhibitor. Examples of HDAC I/II inhibitors include hydroxamic acids, cyclic peptides, benzamides, short-chain fatty acids, and depudecin.
Examples of hydroxamic acids and hydroxamic acid derivatives include, but are not limited to, trichostatin A (TSA), suberoylanilide hydroxamic acid (SAHA), oxamflatin, suberic bishydroxamic acid (SBHA), m-carboxy-cinnamic acid bishydroxamic acid (CBHA), valproic acid and pyroxamide. TSA was isolated as an antifungi antibiotic (Tsuji et al (1976) J. Antibiot (Tokyo) 29:1 -6) and found to be a potent inhibitor of mammalian HDAC (Yoshida et al. (1990) J. Biol. Chem. 265: 17174-17179). The finding that TSA-resistant cell lines have an altered HDAC evidences that this enzyme is an important target for TSA. Other hydroxamic acid- based HDAC inhibitors, SAHA, SBHA, and CBHA are synthetic compounds that are able to inhibit HDAC at micromolar concentration or lower in vitro or in vivo. Glick et al. (1999) Cancer Res. 59:4392-4399. These hydroxamic acid-based HDAC inhibitors all possess an essential structural feature: a polar hydroxamic terminal linked through a hydrophobic methylene spacer (e.g. 6 carbon at length) to another polar site that is attached to a terminal hydrophobic moiety (e.g.. benzene ring). Compounds developed having such essential features also fall within the scope of the hydroxamic acids that may be used as HDAC inhibitors.
Cyclic peptides used as HDAC inhibitors are mainly cyclic tetrapeptides. Examples of cyclic peptides include, but are not limited to, trapoxin A, apicidin and depsipeptide. Trapoxin A is a cyclic tetrapeptide that contains a 2-amino-8-oxo- 9, 10-epoxy-decanoyl (AOE) moiety. Kijima et al. (1993) J. Biol. Chem. 268:22429- 22435. Apicidin is a fungal metabolite that exhibits potent, broad-spectrum antiprotozoal activitity and inhibits HDAC activity at nanomolar concentrations. Darkin-Rattray et al. (1996) Proc. Natl. Acad. Sci. USA. 93; 13143-13147. Depsipeptide is isolated from Chromobacterium violaceum, and has been shown to inhibit HDAC activity at micromolar concentrations.
Examples of benzamides include but are not limited to MS-27-275. Saito et al. (1990) Proc. Natl. Acad. Sci. USA. 96:4592-4597. Examples of short-chain fatty acids include but are not limited to butyrates (e.g., butyric acid, arginine butyrate and phenylbutyrate (PB)). Newmark et al. (1994) Cancer Lett. 78: 1-5; and Carducci et al. (1997) Anticancer Res. 17:3972-3973. In addition, depudecin which has been shown to inhibit HDAC at micromolar concentrations (Kwon et al. (1998) Proc. Natl. Acad. Sci. USA. 95:3356-3361) also falls within the scope of histone deacetylase inhibitors as described herein. Blood Coagulation Disorders
In other aspects, formulations of the invention can be used to treat or prevent blood coagulation disorders (or hemostatic disorders). As used interchangeably herein, the terms "hemostasis", "blood coagulation," and "blood clotting" refer to the control of bleeding, including the physiological properties of vasoconstriction and coagulation. Blood coagulation assists in maintaining the integrity of mammalian circulation after injury, inflammation, disease, congenital defect, dysfunction or other disruption. After initiation of clotting, blood coagulatron proceeds through the sequential activation of certain plasma proenzymes to their enzyme forms (see, for example, Coleman, R. W. et al. (eds.) Hemostasis and Thrombosis, Second Edition, (1987)). These plasma glycoproteins, including Factor XII, Factor XI, Factor IX, Factor X, Factor VII, and prothrombin, are zymogens of serine proteases. Most of these blood clotting enzymes are effective on a physiological scale only when assembled in complexes on membrane surfaces with protein cofactors such as Factor VIII and Factor V. Other blood factors modulate and localize clot formation, or dissolve blood clots. Activated protein C is a specific enzyme that inactivates procoagulant components. Calcium ions are involved in many of the component reactions. Blood coagulation follows either the intrinsic pathway, where all of the protein components are present in blood, or the extrinsic pathway, where the cell-membrane protein tissue factor plays a critical role. Clot formation occurs when fibrinogen is cleaved by thrombin to form fibrin. Blood clots are composed of activated platelets and fibrin.
Further, the formation of blood clots does not only limit bleeding in case of an injury (hemostasis), but may lead to serious organ damage and death in the context of atherosclerotic diseases by occlusion of an important artery or vein. Thrombosis is thus blood clot formation at the wrong time and place. It involves a cascade of complicated and regulated biochemical reactions between circulating blood proteins (coagulation factors), blood cells (in particular platelets), and elements of an injured vessel wall. Accordingly, the present invention provides anticoagulation and antithrombotic treatments aiming at inhibiting the formation of blood clots in order to prevent or treat blood coagulation disorders, such as myocardial infarction, stroke, loss of a limb by peripheral artery disease or pulmonary embolism.
As used interchangeably herein, "modulating or modulation of hemostasis" and "regulating or regulation of hemostasis" includes the induction (e.g., stimulation or increase) of hemostasis, as well as the inhibition (e.g., reduction or decrease) of hemostasis.
In one aspect, the invention provides a method for reducing or inhibiting hemostasis in a subject by administering a formulation of the invention. The compositions and methods disclosed herein are useful for the treatment or prevention of thrombotic disorders. As used herein, the term "thrombotic disorder" includes any disorder or condition characterized by excessive or unwanted coagulation or hemostatic activity, or a hypercoagulable state. Thrombotic disorders include diseases or disorders involving platelet adhesion and thrombus formation, and may manifest as an increased propensity to form thromboses, e.g., an increased number of thromboses, thrombosis at an early age, a familial tendency towards thrombosis, and thrombosis at unusual sites. Examples of thrombotic disorders include, but are not limited to, thromboembolism, deep vein thrombosis, pulmonary embolism, stroke, myocardial infarction, miscarriage, thrombophilia associated with anti-thrombin III deficiency, protein C deficiency, protein S deficiency, resistance to activated protein C, dysfibrinogenemia, fibrinolytic disorders, homocystinuria, pregnancy, inflammatory disorders, myeloproliferative disorders, arteriosclerosis, angina, e.g., unstable angina, disseminated intravascular coagulation, thrombotic thrombocytopenic purpura, cancer metastasis, sickle cell disease, glomerular nephritis, and drug induced thrombocytopenia (including, for example, heparin induced thrombocytopenia). In addition, formulation of the inventions may be administered to prevent thrombotic events or to prevent re-occlusion during or after therapeutic clot lysis or procedures such as angioplasty or surgery.
In another embodiment, a combination drug regimen may include drugs or compounds for the treatment or prevention of blood coagulation disorders or secondary conditions associated with these conditions. Thus, a combination drug regimen may include a formulation of the invention and one or more anticoagulation or anti-thrombosis agents. For example, a formulation of the invention can be combined with an effective amount of one or more of: aspirin, heparin, and oral Warfarin that inhibits Vit K-dependent factors, low molecular weight heparins that inhibit factors X and II, thrombin inhibitors, inhibitors of platelet GP HbIIIa receptors, inhibitors of tissue factor (TF), inhibitors of human von Willebrand factor, inhibitors of one or more factors involved in hemostasis (in particular in the coagulation cascade). In addition, formulations of the invention can be combined with thrombolytic agents, such as t-PA, streptokinase, reptilase, TNK-t-PA, and staphylokinase. Weight Control
In another aspect, a formulation of the invention may be used for treating or preventing weight gain or obesity in a subject. For example, a formulation of the invention may be used, for example, to treat or prevent hereditary obesity, dietary obesity, hormone related obesity, obesity related to the administration of medication, to reduce the weight of a subject, or to reduce or prevent weight gain in a subject. A subject in need of such a treatment may be a subject who is obese, likely to become obese, overweight, or likely to become overweight-Subjects who are likely to become obese or overweight can be identified, for example, based on family history, genetics, diet, activity level, medication intake, or various combinations thereof. In yet other embodiments, a formulation of the invention may be administered to subjects suffering from a variety of other diseases and conditions that may be treated or prevented by promoting weight loss in the subject. Such diseases include, for example, high blood pressure, hypertension, high blood cholesterol, dyslipidemia, type 2 diabetes, insulin resistance, glucose intolerance, hyperinsulinemia, coronary heart disease, angina pectoris, congestive heart failure, stroke, gallstones, cholescystitis and cholelithiasis, gout, osteoarthritis, obstructive sleep apnea and respiratory problems, some types of cancer (such as endometrial, breast, prostate, and colon), complications of pregnancy, poor female reproductive health (such as menstrual irregularities, infertility, irregular ovulation), bladder control problems (such as stress incontinence); uric acid nephrolithiasis; psychological disorders (such as depression, eating disorders, distorted body image, and low self esteem). Stunkard AJ, Wadden TA. (Editors) Obesity: theory and therapy, Second Edition. New York: Raven Press, 1993. Finally, patients with AIDS can develop lipodystrophy or insulin resistance in response to combination therapies for AIDS.
In another embodiment, a formulation of the invention may be used for inhibiting adipogenesis or fat cell differentiation, whether in vitro or in vivo. In particular, high circulating levels of insulin and/or insulin like growth factor (IGF) 1 will be prevented from recruiting preadipocytes to differentiate into adipocytes. Such methods may be used for treating or preventing obesity.
In other embodiments, a formulation of the invention may be used for reducing appetite and/or increasing satiety, thereby causing weight loss or avoidance of weight gain. A subject in need of such a treatment may be a subject who is overweight, obese or a subject likely to become overweight or obese. The method may comprise administering daily or, every other day, or once a week, a dose, e.g., in the form of a pill, to a subject. The dose may be an "appetite reducing dose.'" Also provided are methods for modulating adipogenesis or fat cell differentiation, whether in vitro or in vivo. In particular, high circulating levels of insulin and/or insulin like growth factor (IGF) 1 will be prevented from recruiting preadipocytes to differentiate into adipocytes. Such methods may be used to modulate obesity. A method for stimulating adipogenesis may comprise contacting a cell with a formulation of the invention. A method for modulating weight may further comprise monitoring the weight of the subject and/or the level of modulation of sirtuins, for example, in adipose tissue.
In an exemplary embodiment, a formulation of the invention may be administered as a combination therapy for treating or preventing weight gain or obesity. For example, a formulation of the invention may be administered in combination with one or more anti-obesity agents. Exemplary anti-obesity agents include, for example, phenylpropanolamine, ephedrine, pseudoephedrine, phentermine, a cholecystokinin-A agonist, a monoamine reuptake inhibitor (such as sibutramine), a sympathomimetic agent, a serotonergic agent (such as dexfenfluramine or fenfluramine), a dopamine agonist (such as bromocriptine), a melanocyte-stimulating hormone receptor agonist or mimetic, a melanocyte- stimulating hormone analog, a cannabinoid receptor antagonist, a melanin concentrating hormone antagonist, the OB protein (leptin), a leptin analog, a leptin receptor agonist, a galanin antagonist or a GI lipase inhibitor or decreaser (such as orlistat). Other anorectic agents include bombesin agonists, dehydroepiandiOsterone or analogs thereof, glucocorticoid receptor agonists and antagonists, orexin receptor antagonists, urocortin binding protein antagonists, agonists of the glucagon-like peptide- 1 receptor such as Exendin and ciliary neurotrophic factors such as Axokine. In another embodiment, a formulation of the invention may be administered to reduce drug-induced weight gain. For example, a formulation of the invention - may be administered as a combination therapy with medications that may stimulate appetite or cause weight gain, in particular, weight gain due to factors other than water retention. Examples of medications that may cause weight gain, include for example, diabetes treatments, including, for example, sulfonylureas (such as glipizide and glyburide), thiazolidinediones (such as pioglitazone and rosiglitazone), meglitinides, nateglinide, repaglinide, sulphonylurea medicines, and insulin; antidepressants, including, for example, tricyclic antidepressants (such as amitriptyline and imipramine), irreversible monoamine oxidase inhibitors (MAOIs), selective serotonin reuptake inhibitors (SSRIs), bupropion, paroxetine, and mirtazapine; steroids, such as, for example, prednisone; hormone therapy; lithium carbonate; valproic acid; carbamazepine; chlorpromazine; thiothixene; beta blockers (such as propranolol); alpha blockers (such as clonidine, prazosin and terazosin); and contraceptives including oral contraceptives (birth control pills) or other contraceptives containing estrogen and/or progesterone (Depo-Provera, Norplant, Ortho), testosterone or Megestrol. In another exemplary embodiment, formulations of the invention may be administered as part of a smoking cessation program to prevent weight gain or reduce weight already gained. Metabolic Disorders/Diabetes
In another aspect, formulations of the invention may be used for treating or preventing a metabolic disorder, such as insulin-resistance, a pre-diabetic state, type II diabetes, and/or complications thereof. Administration of a formulation of the invention may increase insulin sensitivity and/or decrease insulin levels in a subject. A subject in need of such a treatment may be a subject who has insulin resistance or other precursor symptom of type II diabetes, who has type II diabetes, or who is likely to develop any of these conditions. For example, the subject may be a subject having insulin resistance, e.g., having high circulating levels of insulin and/or associated conditions, such as hyperlipidemia, dyslipogenesis, hypercholesterolemia, impaired glucose tolerance, high blood glucose sugar level, other manifestations of syndrome X, hypertension, atherosclerosis and lipodystrophy.
In an exemplary embodiment, formulations of the invention may be administered as a combination therapy for treating or preventing a metabolic disorder. For example, a formulation of the invention may be administered in combination with one or more anti-diabetic agents. Exemplary anti-diabetic agents include, for example, an aldose reductase inhibitor, a glycogen phosphorylase inhibitor, a sorbitol dehydrogenase inhibitor, a protein tyrosine phosphatase 1 B inhibitor, a dipeptidyl protease inhibitor, insulin (including orally bioavailable insulin preparations), an insulin mimetic, metformin, acarbose, a peroxisome proliferator-activated receptor-γ (PPAR-γ) ligand such as troglitazone, rosaglitazone, pioglitazone or GW- 1929, a sulfonylurea, glipazide, glyburide, or - chlorpropamide wherein the amounts of the first and second compounds result in a therapeutic effect. Other anti-diabetic agents include a glucosidase inhibitor, a glucagon-like peptide- 1 (GLP-I ), insulin, a PPAR α/γ dual agonist, a meglitimide and an αP2 inhibitor. In an exemplary embodiment, an anti-diabetic agent may be a dipeptidyl peptidase IV (DP-IV or DPP-IV) inhibitor, such as, for example LAF237 from Novartis (NVP DPP728; l -[[[2-[(5-cyanopyridin-2-yl)amino] ethyl]amino]acetyl]-2- cyano-(S)- pyrrolidine) or MK-04301 from Merck (see e.g., Hughes et al., Biochemistry 38: 1 1597-603 (1999)). Inflammatory Diseases
In other aspects, formulations of the invention can be used to treat or prevent a disease or disorder associated with inflammation. Formulations of the invention may be administered prior to the onset of, at, or after the initiation of inflammation. When used prophylactically, the formulations of the invention are preferably provided in advance of any inflammatory response or symptom.
Administration of the compounds may prevent or attenuate inflammatory responses or symptoms.
Exemplary inflammatory conditions include, for example, multiple sclerosis, rheumatoid arthritis, psoriatic arthritis, degenerative joint disease, spondouloarthropathies, gouty arthritis, systemic lupus erythematosus, juvenile arthritis, rheumatoid arthritis, osteoarthritis, osteoporosis, diabetes (e.g., insulin dependent diabetes mellitus or juvenile onset diabetes), menstrual cramps, cystic fibrosis, inflammatory bowel disease, irritable bowel syndrome, Crohn's disease, mucous colitis, ulcerative colitis, gastritis, esophagitis, pancreatitis, peritonitis, Alzheimer's disease, shock, ankylosing spondylitis, gastritis, conjunctivitis, pancreatis (acute or chronic), multiple organ injury syndrome (e.g., secondary to septicemia or trauma), myocardial infarction, atherosclerosis, stroke, reperfusion injury (e.g., due to cardiopulmonary bypass or kidney dialysis), acute glomerulonephritis, vasculitis, thermal injury (i.e., sunburn), necrotizing enterocolitis, granulocyte transfusion associated syndrome, and/or Sjogren's syndrome. Exemplary inflammatory conditions of the skin include, for example, eczema, atopic dermatitis, contact dermatitis, urticaria, scleroderma, psoriasis, and dermatosis with acute inflammatory components.
In another embodiment, a formulation of the invention may be used to treat or prevent allergies and respiratory conditions, including asthma, bronchitis, pulmonary fibrosis, allergic rhinitis, oxygen toxicity, emphysema, chronic bronchitis, acute respiratory distress syndrome, and any chronic obstructive pulmonary disease (COPD). The formulation of the invention may be used to treat chronic hepatitis infection, including hepatitis B and hepatitis C.
Additionally, a formulation of the invention may be used to treat autoimmune diseases and/or inflammation associated with autoimmune diseases such as organ-tissue autoimmune diseases (e.g., Raynaud's syndrome), scleroderma, myasthenia gravis, transplant rejection, endotoxin shock, sepsis, - psoriasis, eczema, dermatitis, multiple sclerosis, autoimmune thyroiditis, uveitis, systemic lupus erythematosis, Addison's disease, autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome), and Grave's disease. In certain embodiments, a formulation of the invention may be taken alone or in combination with other compounds useful for treating or preventing inflammation. Exemplary anti-inflammatory agents include, for example, steroids (e.g., Cortisol, cortisone, fludrocortisone, prednisone, 6α-methylprednisone, triamcinolone, betamethasone or dexamethasone), nonsteroidal antiinflammatory drugs (NSAIDS (e.g., aspirin, acetaminophen, tolmetin, ibuprofen, mefenamic acid, piroxicam, nabumetone, rofecoxib, celecoxib, etodolac or nimesulide). In another embodiment, the other therapeutic agent is an antibiotic (e.g., vancomycin, penicillin, amoxicillin, ampicillin, cefotaxime, ceftriaxone, cefixime, rifampinmetronidazole, doxycycline or streptomycin). In another embodiment, the other therapeutic agent is a PDE4 inhibitor (e.g., roflumilast or rolipram). In another embodiment, the other therapeutic agent is an antihistamine (e.g., cyclizine, hydroxyzine, promethazine or diphenhydramine). In another embodiment, the other therapeutic agent is an anti-malarial (e.g., artemisinin, artemether, artsunate, chloroquine phosphate, mefloquine hydrochloride, doxycycline hyclate, proguanil hydrochloride, atovaquone or halofantrine). In one embodiment, the other therapeutic agent is drotrecogin alfa.
Further examples of anti-inflammatory agents include, for example, aceclofenac, acemetacin, e-acetamidocaproic acid, acetaminophen, acetaminosalol, acetanilide, acetylsalicylic acid, S-adenosylmethioninc, alclofenac, alclometasone, alfentanil, algestone, allylprodine, alminoprofen; aloxiprin, alphaprodine, aluminum bis(acetylsalicylate), amcinonide, amfenac, aminochlorthenoxazin, 3- amino-4-hydroxybutyric acid, -2-amino-4-picoline, aminopropylon, aminopyrine, amixetrine, ammonium salicylate, ampiroxicam, amtolmetin guacil, anileridine, antipyrine, antrafenine, apazone, beclomethasone, bendazac, benorylate, benoxaprofen, benzpiperylon, benzydamine, benzylmoφhine, bermoprofen, betamethasone, betamethasone- 17-valerate, bezitramide, α-bisabolol, bromfenac, p-bromoacetanilide, 5-bromosalicylic acid acetate, bromosaligenin, bucetin, bucloxic acid, bucolόme, budesonide, bufexamac, bumadizon, buprenoφhine, butacetin, butibufen, butorphanol, carbamazepine, carbiphene, carprofen, carsalam, chlorobutanol, chloroprednisone, chlorthenoxazin, choline salicylate, cinchophen, cinmetacin, ciramadol, clidanac, clobetasol, clocortolone, clometacin, clonitazene, clonixin, clopirac, cloprednol, clove, codeine, codeine methyl bromide, codeine phosphate, codeine sulfate, cortisone, cortivazol, cropropamide, crotethamide, cyclazocine, deflazacort, dehydrotestosterone, desomorphine, desonide, desoximetasone, dexamethasone, dexamethasone-21-isonicotinate, dexoxadrol, dextromoramide, dextropropoxyphene, deoxycorticosterone, dezocine, diampromide, diamorphone, diclofenac, difenamizole, difenpiramide, diflorasone, diflucortolone, diflunisal, difluprednate, dihydrocodeine, dihydrocodeinone enol acetate, dihydromorphine, dihydroxyaluminum acetylsalicylate, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetyl butyrate, dipipanone, diprocetyl, dipyrone, ditazol, droxicam, emorfazone, enfenamic acid, enoxolone, epirizole, eptazocine, etersalate, ethenzamide, ethoheptazine, ethoxazene, ethylmethylthiambutene, ethylmorphine, etodolac, etofenamate, etonitazene, eugenol, felbinac, fenbufen, fenclozic acid, fendosal, fenoprofen, fentanyl, fentiazac, fepradinol, feprazone, floctafenine, fluazacort, flucloronide, flufenamic acid, flumethasone; flunisolide, flunixin, flunoxaprofen, fluocinolone acetonide, fluocinonide, fluocinolone acetonide, fluocortin butyl, tluocortolone, fluoresone, fluorometholone, fluperolone, flupirtine, fluprednidene, fluprednisolone, fluproquazone, flurandrenolide, flurbiprofen, fluticasone, formocortal, fosfosal, gentisic acid, glafenine, glucametacin, glycol salicylate, guaiazulene, halcinonide, halobetasol, halometasone, haloprednone, heroin, hydrocodone, hydrocortamate, hydrocortisone, hydrocortisone acetate, hydrocortisone succinate, hydrocortisone hemisuccinate, hydrocortisone 21 -lysinate, hydrocortisone cypionate, hydromoφhone, hydroxypethidine, ibufenac, ibuprofen, ibuproxam, imidazole salicylate, indomethacin, indoprofen, isofezolac, isoflupredone, isoflupredone acetate, isoladol, isomethadone, isonixin, isoxepac, isoxicam, ketobemidone, ketoprofen, ketorolac, p-lactophenetide, lefetamine, levallorphan, levorphanol, levophenacyl-moφhan, lofentanil, lonazolac, lornoxicam, loxoprofen, lysine acetylsalicylate, mazipredone, meclofenamic acid, medrysone, mefenamic acid, meloxicam, meperidine, meprednisone, meptazinol, mesalamine, metazocine, methadone, methotrimeprazine, methylprednisolone, methylprednisolone acetate, methylprednisolone sodium succinate, methylprednisolone suleptnate, metiazinic acid, metofoline, metopon, mofebutazone, mofezolac, mometasone, morazone, morphine, morphine hydrochloride, morphine sulfate, morpholine salicylate, myrophine, nabumetone, nalbuphine, nalorphine, 1 -naphthyl salicylate, naproxen, narceine, nefopam, nicomorphine, nifenazone, niflumic acid, nimesulide, 5'-nitro- 2'-propoxyacetanilide, norlevoφhanol, normethadone, normoφhine, norpipanone, olsalazine, opium, oxaceprol, oxametacine, oxaprozin, oxycodone, oxymorphone, oxyphenbutazone, papaveretum, paramethasone, paranyline, parsalmide, pentazocine, perisoxal, phenacetin, phenadoxone, phenazocine, phenazopyridine hydrochloride, phenocoll, phenoperidine, phenopyrazone, phenomoφhan, phenyl acetylsalicylate, phenylbutazone, phenyl salicylate, phenyramidol, piketoprofen, piminodine, pipebuzone, piperylone, pirazolac, piritramide, piroxicam, piφrofen, pranoprofen, prednicarbate, prednisolone, prednisone, prednival, prednylidene, proglumetacin, proheptazine, promedol, propacetamol, properidine, propiram, propoxyphene, propyphenazone, proquazone, protizinic acid, proxazole, ramifenazone, remifentanil, rimazolium metilsulfate, salacetamide, salicin, salicylamide^ salicylamide o-acetic acid, salicylic acid, salicylsulfuric acid, salsalate, salverine, simetride, sufentanil, sulfasalazine, sulindac, superoxide dismutase, suprofen, suxibuzone, talniflumate, tenidap, tenoxicam, terofenamate, tetrandrine, thiazolinobutazone, tiaprofenic acid, tiaramide, tilidine, tinoridine, tixocortol, tolfenamic acid, tolmetin, tramadol, triamcinolone, triamcinolone acetonide, tropesin, viminol, xenbucin, ximoprofen, zaltoprofen and zomepirac. In an exemplary embodiment, a formulation of the invention may be administered with a selective COX-2 inhibitor for treating or preventing inflammation. Exemplary selective COX-2 inhibitors include, for example, deracoxib, parecoxib, celecoxib, valdecoxib, rofecoxib, etoricoxib, lumiracoxib, 2- (3,5-difluorophenyl)-3-[4-(methylsulfonyl)phenyl]-2-cyclopenten-l -one, (S)-6,8- dichloro-2-(triflu- oromethyl)-2H-l -benzopyran-3-carboxylic acid, 2-(3,4- difluorophenyl)-4-(3-hydroxy-3-methyl-l-butoxy)-5-[4-(methylsulfonyl)phenyl]-3- (2H)-pyridazinone, 4-[5-(4-fluorophenyl)-3-(trifluoromethyl)-l H-pyrazol-1 - yljbenzenesulfonamide, tert-butyl 1 benzyl-4-[(4-oxopiperidin-l- yl}sulfonyl]piperidine-4-carboxylate, 4-[5-(phenyl)-3-(trifluoromethyl)-lH- pyrazol-1 -yljbenzenesulfonamide, salts and prodrugs thereof. Flushing In another aspect, formulations of the invention may be used for reducing the incidence or severity of flushing and/or hot flashes which are symptoms of a disorder. For instance, the subject method includes the use of formulations of the invention, alone or in combination with other agents, for reducing incidence or severity of flushing and/or hot flashes in cancer patients. In other embodiments, the method provides for the use of formulations of the invention to reduce the incidence or severity of flushing and/or hot flashes in menopausal and post-menopausal woman.
In another aspect, formulations of the invention may be used as a therapy for reducing the incidence or severity of flushing and/or hot flashes which are side.- effects of another drug therapy, e.g., drug-induced flushing. In certain embodiments, a method for treating and/or preventing drug-induced flushing comprises administering to a patient in need thereof a formulation comprising at least one flushing inducing compound and a formulation of the invention. In other embodiments, a method for treating drug induced flushing comprises separately administering one or more compounds that induce flushing and a formulation of the invention, e.g., wherein the formulation of the invention and flushing inducing agent have not been formulated in the same compositions. When using separate formulations, the formulation of the invention may be administered ( 1 ) at the same as administration of the flushing inducing agent, (2) intermittently with the flushing inducing agent, (3) staggered relative to administration of the flushing inducing agent, (4) prior to administration of the flushing inducing agent, (5) subsequent to administration of the flushing inducing agent, and (6) various combination thereof. Exemplary flushing inducing agents include, for example, niacin, faloxifene, antidepressants, anti-psychotics, chemotherapeutics, calcium channel blockers, and antibiotics.
In one embodiment, formulations of the invention may be used to reduce flushing side effects of a vasodilator or an antilipemic agent (including anticholesteremic agents and lipotropic agents). In an exemplary embodiment, a formulation of the invention may be used to reduce flushing associated with the administration of niacin.
Nicotinic acid, 3-pyridinecarboxylic acid or niacin, is an antilipidemic agent that is marketed under, for example, the trade names Nicolar®, SloNiacin®, Nicobid® and Time Release Niacin®. Nicotinic acid has been used for many years in the treatment of lipidemic disorders such as hyperlipidemia, hypercholesterolemia and atherosclerosis. This compound has long been known to exhibit the beneficial effects of reducing total cholesterol, low density lipoproteins or "LDL cholesterol," triglycerides and apolipoprotein a (Lp(a)) in the human body, while increasing desirable high density lipoproteins or "HDL cholesterol".
Typical doses range from about 1 gram to about 3 grams daily. Nicotinic acid is normally administered two to four times per day after meals, depending upon the dosage form selected. Nicotinic acid isxurrently commercially available in two dosage forms. One dosage form is an immediate or rapid release tablet which should be administered three or four times per day. Immediate release ("IR") nicotinic acid formulations generally release nearly all of their nicotinic acid within about 30 to 60 minutes following ingestion. The other dosage form is a sustained release form which is suitable for administration two to four times per day. In contrast to IR formulations, sustained release ("SR") nicotinic acid formulations are designed to release significant quantities of drug for absorption into the blood stream over specific timed intervals in order to maintain therapeutic levels of nicotinic acid over an extended period such as 12 or 24 hours after ingestion.
As used herein, the term "nicotinic acid'* is meant to encompass nicotinic acid or a compound other than nicotinic acid itself which the body metabolizes into nicotinic acid, thus producing essentially the same effect as nicotinic acid. Exemplary compounds that produce an effect similar to that of nicotinic acid include, for example, nicotinyl alcohol tartrate, d-glucitol hexanicotinate, aluminum nicotinate, niceritrol and d, 1 -alpha-tocopheryl nicotinate. Each such compound will be collectively referred to herein as "nicotinic acid."
In another embodiment, the invention provides a method for treating and/or preventing hyperlipidemia with reduced flushing side effects. The method comprises the steps of administering to a subject in need thereof a therapeutically effective amount of nicotinic acid and a formulation of the invention in an amount sufficient to reduce flushing. In an exemplary embodiment, the nicotinic acid and/or formulation of the invention may be administered nocturnally. In another representative embodiment, the method involves the use of formulations of the invention to reduce flushing side effects of raloxifene. Raloxifene acts like estrogen in certain places in the body, but is not a hormone. It helps prevent osteoporosis in women who have reached menopause. Osteoporosis causes bones to gradually grow thin, fragile, and more likely to break. Evista slows down the loss of bone mass that occurs with menopause, lowering the risk of spine fractures due to osteoporosis. A common side effect of raloxifene is hot flashes (sweating and flushing). This can be uncomfortable for women who already have hot flashes due to menopause.
In another representative embodiment, the method involves the use of formulations of the invention to reduce flushing side effects of antidepressants or anti-psychotic agent. For instance, formulations of the invention can be used in conjunction (administered separately or together) with a serotonin reuptake . inhibitor, a 5HT2 receptor antagonist, an anticonvulsant, a norepinephrine reuptake inhibitor, an α-adrenoreceptor antagonist, an NK-3 antagonist, an NK- I receptor antagonist, a PDE4 inhibitor, an Neuropeptide Y5 Receptor Antagonists, a D4 receptor antagonist, a 5HTl A receptor antagonist, a 5HTl D receptor antagonist, a CRF antagonist, a monoamine oxidase inhibitor, or a sedative-hypnotic drug.
In certain embodiments, formulations of the invention may be used as part of a treatment with a serotonin reuptake inhibitor (SRI) to reduce flushing. In certain preferred embodiments, the SRI is a selective serotonin reuptake inhibitor (SSRI), such as a fluoxetinoid (fluoxetine, norfluoxetine) or a nefazodoiloid (nefazodone, hydroxynefazodone, oxonefazodone). Other exemplary SSRI's include duloxetine, venlafaxine, milnacipran, citalopram, fluvoxamine, paroxetine and sertraline. The formulation of the invention can also be used as part of a treatment with sedative- hypnotic drug, such as selected from the group consisting of a benzodiazepine (such as alprazolam, chlordiazepoxide, clonazepam, chlorazepate, clobazam, diazepam, halazepam, lorazepam, oxazepam and prazepam), Zolpidem, and barbiturates. In still other embodiments, a formulation of the invention may be used as part of a treatment with a 5-HT1 A receptor partial agonist, such as selected from the group consisting of buspirone, flesinoxan, gepirone and ipsapirone. Formulations of the invention can also used as part of a treatment with a norepinephrine reuptake inhibitor, such as selected from tertiary amine tricyclics and secondary amine . tricyclics. Exemplary tertiary amine tricyclics include amitriptyline, clomipramine, doxepin, imipramine and trimipramine. Exemplary secondary amine tricyclics include amoxapine, desipramine, maprotiline, nortriptyline and protriptyline. In certain embodiments, formulations of the invention may be used as part of a treatment with a monoamine oxidase inhibitor, such as selected from the group consisting of isocarboxazid, phenelzine, tranylcypromine, selegiline and moclobemide.
In still another representative embodiment, formulations of the invention may be used to reduce flushing side effects of chemotherapeutic agents, such as cyclophosphamide, tamoxifen.
In another embodiment, formulations of the invention may be used to reduce flushing side effects of calcium channel blockers, such as amlodipine.
In another embodiment, formulations of the invention may be used to reduce flushing side effects of antibiotics. For example, formulations of the invention can be used in combination with levofloxacin. Levofloxacin is used to treat infections of the sinuses, skin, lungs, ears, airways, bones, and joints caused by susceptible bacteria. Levofloxacin also is frequently used to treat urinary infections, including - those resistant to other antibiotics, as well as prostatitis. Levofloxacin is effective in treating infectious diarrheas caused by E. coli, Campylobacter jejuni, and shigella bacteria. Levofloxacin also can be used to treat various obstetric infections, including mastitis. Ocular Disorders One aspect of the present invention is a method for inhibiting, reducing or otherwise treating vision impairment by administering to a patient a therapeutic dosage of a formulation of the invention.
In certain aspects of the invention, the vision impairment is caused by damage to the optic nerve or central nervous system. In particular embodiments, optic nerve damage is caused by high intraocular pressure, such as that created by glaucoma. In other particular embodiments, optic nerve damage is caused by swelling of the nerve, which is often associated with an infection or an immune (e.g., autoimmune) response such as in optic neuritis. Glaucoma describes a group of disorders which are associated with a visual field defect, cupping of the optic disc, and optic nerve damage. These are commonly referred to as glaucomatous optic neuropathies. Most glaucomas are usually, but not always, associated with a rise in intraocular pressure. Exemplary forms of glaucoma include Glaucoma and Penetrating Keratoplasty, Acute Angle Closure, Chronic Angle Closure, Chronic Open Angle, Angle Recession, Aphakic and Pseudophakic, Drug-Induced, Hyphema, Intraocular Tumors, Juvenile, Lens-Particle, Low Tension, Malignant, Neovascular, Phacolytic, Phacomorphic, Pigmentary, Plateau Iris, Primary Congenital, Primary Open Angle, Pseudoexfoliation, Secondary Congenital, Adult Suspect, Unilateral, Uveitic, Ocular Hypertension, Ocular Hypotony, Posner-Schlossman Syndrome and Scleral Expansion Procedure in Ocular Hypertension & Primary Open-angle Glaucoma.
Intraocular pressure can also be increased by various surgical procedures, such as phacoemulsification (i.e., cataract surgery) and implanation of structures such as an artificial lens. In addition, spinal surgeries in particular, or any surgery in which the patient is prone for an extended period of time can lead to increased interoccular pressure.
Optic neuritis (ON) is inflammation of the optic nerve and causes acute loss of vision. It is highly associated with multiple sclerosis (MS) as 15-25% of MS patients initially present with ON, and 50-75% of ON patients are diagnosed with MS. ON is also associated with infection (e.g., viral infection, meningitis, syphilis), inflammation (e.g., from a vaccine), infiltration and ischemia. Another condition leading to optic nerve damage is anterior ischemic optic neuropathy (AION). There are two types of AION. Arteritic AION is due to giant cell arteritis (vasculitis) and leads to acute vision loss. Non-arteritic AION encompasses all cases of ischemic optic neuropathy other than those due to giant cell arteritis. The pathophysiology of AION is unclear although it appears to incorporate both inflammatory and ischemic mechanisms.
Other damage to the optic nerve is typically associated with demyleination, inflammation, ischemia, toxins, or trauma to the optic nerve. Exemplary conditions where the optic nerve is damaged include Demyelinating Optic Neuropathy (Optic Neuritis, Retrobulbar Optic Neuritis), Optic Nerve Sheath Meningioma, Adult Optic Neuritis, Childhood Optic Neuritis, Anterior Ischemic Optic Neuropathy, Posterior Ischemic Optic Neuropathy, Compressive Optic Neuropathy, Papilledema, Pseudopapilledema and Toxic/Nutritional Optic Neuropathy.
Other neurological conditions associated with vision loss, albeit not directly associated with damage to the optic nerve, include Amblyopia, Bells Palsy, Chronic Progressive External Ophthalmoplegia, Multiple Sclerosis, Pseudotumor Cerebri and Trigeminal Neuralgia.
In certain aspects of the invention, the vision impairment is caused by retinal damage. In particular embodiments, retinal damage is caused by disturbances in blood flow to the eye (e.g., arteriosclerosis, vasculitis). In particular embodiments, retinal damage is caused by disrupton of the macula (e.g., exudative or non- exudative macular degeneration).
Exemplary retinal diseases include Exudative Age Related Macular Degeneration, Nonexudative Age Related Macular Degeneration, Retinal Electronic Prosthesis and RPE Transplantation Age Related Macular Degeneration, Acute Multifocal Placoid Pigment Epitheliopathy, Acute Retinal Necrosis, Best Disease, Branch Retinal Artery Occlusion, Branch Retinal Vein Occlusion, Cancer Associated and Related Autoimmune Retinopathies, Central Retinal Artery Occlusion, Central Retinal Vein Occlusion, Central Serous Chorioretinopathy, Eales Disease, Epimacular Membrane, Lattice Degeneration, Macroaneurysm, Diabetic Macular Edema, Irvine-Gass Macular Edema, Macular Hole, Subretinal Neovascular Membranes, Diffuse Unilateral Subacute Neuroretinitis, Nonpseudophakic Cystoid Macular Edema, Presumed Ocular Histoplasmosis Syndrome, Exudative Retinal Detachment, Postoperative Retinal Detachment, Proliferative Retinal Detachment, Rhegmatogenous Retinal Detachment, Tractional Retinal Detachment, Retinitis Pigmentosa, CMV Retinitis, Retinoblastoma, Retinopathy of Prematurity, Birdshot Retinopathy, Background Diabetic
Retinopathy, Proliferative Diabetic Retinopathy, Hemoglobinopathies Retinopathy, Purtscher Retinopathy, Valsalva Retinopathy, Juvenile Retinoschisis, Senile Retinoschisis, Terson Syndrome and White Dot Syndromes.
Other exemplary diseases include ocular bacterial infections (e.g. conjunctivitis, keratitis, tuberculosis, syphilis, gonorrhea), viral infections (e.g. Ocular Herpes Simplex Virus, Varicella Zoster Virus, Cytomegalovirus retinitis, Human Immunodeficiency Virus (HIV)) as well as progressive outer retinal necrosis secondary to HIV or other HIV-associated and other immunodeficiency-associated ocular diseases. In addition, ocular diseases include fungal infections (e.g. Candida choroiditis, histoplasmosis), protozoal infections (e.g. toxoplasmosis) and others such as ocular toxocariasis and sarcoidosis.
One aspect of the invention is a method for inhibiting, reducing or treating vision impairment in a subject undergoing treatment with a chemotherapeutic drug (e.g., a neurotoxic drug, a drug that raises intraocular pressure such as a steroid), by administering to the subject in need of such treatment a therapeutic dosage of a formulation of the invention.
Another aspect of the invention is a method for inhibiting, reducing or treating vision impairment in a subject undergoing surgery, including ocular or other surgeries performed in the prone position such as spinal cord surgery, by administering to the subject in need of such treatment a therapeutic dosage of a formulation of the invention. Ocular surgeries include cataract, iridotomy and lens replacements.
Another aspect of the invention is the treatment, including inhibition and prophylactic treatment, of age related ocular diseases include cataracts, dry eye, retinal damage and the like, by administering to the subject in need of such treatment a therapeutic dosage of a formulation of the invention. The formation of cataracts is associated with several biochemical changes in the lens of the eye, such as decreased levels of antioxidants ascorbic acid and glutathione, increased lipid, amino acid and protein- oxidation, increased sodium and calcium, loss of amino acids and decreased lens metabolism. The lens, which lacks blood vessels, is suspended in extracellular fluids in the anterior part of the eye.
Nutrients, such as ascorbic acid, glutathione, vitamin E, selenium, bioflavonoids and carotenoids are required to maintain the transparency of the lens. Low levels of selenium results in an increase of free radical-inducing hydrogen peroxide, which is neutralized by the selenium-dependent antioxidant enzyme glutathione peroxidase. Lens-protective glutathione peroxidase is also dependent on the amino acids methionine, cysteine, glycine and glutamic acid.
Cataracts can also develop due to an inability to properly metabolize galactose found in dairy products that contain lactose, a disaccharide composed of the monosaccharide galactose and glucose. Cataracts can be prevented, delayed, slowed and possibly even reversed if detected early and metabolically corrected.
Retinal damage is attributed, inter alia, to free radical initiated reactions in glaucoma, diabetic retinopathy and age-related macular degeneration (AMD). The eye is a part of the central nervous system and has limited regenerative capability. The retina is composed of numerous nerve cells which contain the highest concentration of polyunsaturated fatty acids (PFA) and subject to oxidation. Free radicals are generated by UV light entering the eye and mitochondria in the rods and cones, which generate the energy necessary to transform light into visual impulses. Free radicals cause peroxidation of the PFA by hydroxyl or superoxide radicals which in turn propagate additional free radicals. The free radicals cause temporary or permanent damage to retinal tissue.
Glaucoma is usually viewed as a disorder that causes an elevated intraocular pressure (lOP) that results in permanent damage to the retinal nerve fibers, but a sixth of all glaucoma cases do not develop an elevated IOP. This disorder is now perceived as one of reduced vascular perfusion and an increase in neurotoxic factors. Recent studies have implicated elevated levels of glutamate, nitric oxide and peroxynitirite in the eye as the causes of the death of retinal ganglion cells. Neuroprotective agents may be the future of glaucoma care. For example, nitric oxide synthase inhibitors block the formation of peroxynitrite from nitric oxide and superoxide. In a recent study, animals treated with aminoguanidine, a nitric oxide synthase inhibitor, had a reduction in the loss of retinal ganglion cells. It was concluded that nitric oxide in the eye caused cytotoxicity in many tissues and neurotoxicity in the central nervous system.
Diabetic retinopathy occurs when the underlying blood vessels develop microvascular abnormalities consisting primarily of microaneurysms and intraretinal hemorrhages. Oxidative metabolites are directly involved with the pathogenesis of diabetic retinopathy and free radicals augment the generation of growth factors that lead to enhanced proliferative activity. Nitric oxide produced by endothelial cells of the vessels may also cause smooth muscle cells to relax and result in vasodilation of segments of the vessel. Ischemia and hypoxia of the retina occur after thickening of the arterial basement membrane, endothelial proliferation and loss of pericytes. The inadequate oxygenation causes capillary obliteration or nonperfusion, arteriolar- venular shunts, sluggish blood flow and an impaired ability of RBCs to release oxygen. Lipid peroxidation of the retinal tissues also occurs as a result of free radical damage.
The macula is responsible for our acute central vision and composed of light- sensing cells (cones) while the underlying retinal pigment epithelium (RPE) and choroid nourish and help remove waste materials. The RPE nourishes the cones with the vitamin A substrate for the photosensitive pigments and digests the cones shed outer tips. RPE is exposed to high levels of UV radiation, and secretes factors that inhibit angiogenesis. The choroid contains a dense vascular network that provides nutrients and removes the waste materials. In AMD, the shed cone tips become indigestible by the RPE, where the cells swell and die after collecting too much undigested material. Collections of undigested waste material, called drusen, form under the RPE. Photoxic damage also causes the accumulation of lipofuscin in RPE cells. The intracellular lipofuscin and accumulation of drusen in Bruch's membrane interferes with the transport of oxygen and nutrients to the retinal tissues, and ultimately leads to RPE and photoreceptor dysfunction. In exudative AMD, blood vessels grow from the choriocapillaris through defects in Bruch's membrane and may grow under the RPE, detaching it from the choroid, and leaking fluid or bleeding.
Macular pigment, one of the protective factors that prevent sunlight from damaging the retina, is formed by the accumulation of nutritionally derived carotenoids, such as lutein, the fatty yellow pigment that serves as a delivery vehicle for other important nutrients and zeaxanthin. Antioxidants such as vitamins C and E, beta-carotene and lutein, as well as zinc, selenium and copper, are all found in the healthy macula. In addition to providing nourishment, these antioxidants protect against free radical damage that initiates macular degeneration. Another aspect of the invention is the prevention or treatment of damage to the eye caused by stress, chemical insult or radiation, by administering to the subject in need of such treatment a therapeutic dosage of a formulation of the invention. Radiation or electromagnetic damage to the eye can include that caused by CRT's or exposure to sunlight or UV. In one embodiment, a combination drug regimen may include drugs or compounds for the treatment or prevention of ocular disorders or secondary conditions associated with these conditions. Thus, a combination drug regimen may include a formulation of the invention and one or more therapeutic agents for the treatment of an ocular disorder. For example, a formulation of the invention can be combined with an effective amount of one or more of: an agent that reduces intraocular pressure, an agent for treating glaucoma, an agent for treating optic - neuritis, an agent for treating CMV Retinopathy, an agent for treating multiple sclerosis, and/or an antibiotic, etc.
In one embodiment, a formulation of the invention can be administered in conjunction with a therapy for reducing intraocular pressure. One group of therapies involves blocking aqueous production. For example, topical beta-adrenergic antagonists (timolol and betaxolol) decrease aqueous production. Topical timolol causes IOP to fall in 30 minutes with peak effects in 1 -2 hours. A reasonable regimen is Timoptic 0.5%, one drop every 30 minutes for 2 doses. The carbonic anhydrase inhibitor, acetazolamide, also decreases aqueous production and should be given in conjunction with topical beta-antagonists. An initial dose of 500 mg is administered followed by 250 mg every 6 hours. This medication may be given orally, intramuscularly, or intravenously. In addition, alpha 2-agonists (e.g., Apraclonidine) act by decreasing aqueous production. Their effects are additive to topically administered beta-blockers. They have been approved for use in controlling an acute rise in pressure following anterior chamber laser procedures, but has been reported effective in treating acute closed-angle glaucoma. A reasonable regimen is 1 drop every 30 minutes for 2 doses.
A second group of therapies for reducing intraocular pressure involve reducing vitreous volume. Hyperosmotic agents can be used to treat an acute attack. These agents draw water out of the globe by making the blood hyperosmolar. Oral glycerol in a dose of 1 mL/kg in a cold 50% solution (mixed with lemon juice to make it more palatable) often is used. Glycerol is converted to glucose in the liver; persons with diabetes may need additional insulin if they become hyperglycemic after receiving glycerol. Oral isosorbide is a metabolically inert alcohol that also can be used as an osmotic agent for patients with acute angle-closure glaucoma. Usual dose is 100 g taken p.o. (220 cc of a 45% solution). This inert alcohol should not be confused with isosorbide dinitrate, a nitrate-based cardiac medication used for angina and for congestive heart failure. Intravenous mannitol in a dose of 1 .0- 1.5 mg/kg also is effective and is well tolerated in patients with nausea and vomiting. These hyperosmotic agents should be used with caution in any patient with a history of congestive heart failure.
A third group of therapies involve facilitating aqueous outflow from the eye. Miotic agents pull the iris from the iridocorneal angle and may help to relieve the obstruction of the trabecular meshwork by the peripheral iris. Pilocarpine 2% (blue eyes)-4% (brown eyes) can be administered every 15 minutes for the first 1 -2 hours. More frequent administration or higher doses may precipitate a systemic cholinergic crisis. NSAIDS are sometimes used to reduce inflammation.
Exemplary therapeutic agents for reducing intraocular pressure include ALPHAGAN® P (Allergan) (brimonidine tartrate ophthalmic solution), AZOPT© (Alcon) (brinzolamide ophthalmic suspension), BETAGAN® (Allergan) (levobunolol hydrochloride ophthalmic solution, USP), BETIMOL© (Vistakon) (timolol ophthalmic solution), BETOPTlC S® (Alcon) (betaxolol HCl), BRIMONIDINE TARTRATE (Bausch & Lomb), CARTEOLOL HYDROCHLORIDE (Bausch & Lomb), COSOPT® (Merck) (dorzolamide hydrochloride-timolol maleate ophthalmic solution), LUMIGAN® (Allergan) (bimatoprost ophthalmic solution), OPTIPRANOLOL® (Bausch & Lomb) (metipranolol ophthalmic solution), TIMOLOL GFS (Falcon) (timolol maleate ophthalmic gel forming solution), TIMOPTIC® (Merck) (timolol maleate ophthalmic solution), TRAVATAN® (Alcon) (travoprost ophthalmic solution), TRUSOPT® (Merck) (dorzolamide hydrochloride ophthalmic solution) and XALATAN® (Pharmacia & Upjohn) (latanoprost ophthalmic solution).
In one embodiment, a formulation of the invention can be administered in conjunction with a therapy for treating and/or preventing glaucoma. An example of a glaucoma drug is DARANIDE® Tablets (Merck) (Dichlorphenamide).
In one embodiment, a formulation of the invention can be administered in conjunction with a therapy for treating and/or preventing optic neuritis. Examples of drugs for optic neuritis include DECADRON® Phosphate Injection (Merck) (Dexamethasone Sodium Phosphate), DEPO-MEDROL® (Pharmacia &
Upjohn)(methylprednisolone acetate), HYDROCORTONE® Tablets (Merck) (Hydrocortisone), ORAPRED® (Biomarin) (prednisolone sodium phosphate oral solution) and PEDIAPRED® (Celltech) (prednisolone sodium phosphate, USP). In one embodiment, a formulation of the invention can be administered in conjunction with a therapy for treating and/or preventing CMV Retinopathy.
Treatments for CMV retinopathy include CYTOVENE® (ganciclovir capsules) and VALCYTE® (Roche Laboratories) (valganciclovir hydrochloride tablets).
In one embodiment, a formulation of the invention can be administered in conjunction with a therapy for treating and/or preventing multiple sclerosis. Examples of such drugs include DANTRIUM® (Procter & Gamble
Pharmaceuticals) (dantrolene sodium), NOVANTRONE® (Serono) (mitoxantrone), AVONEX® (Biogen Idee) (Interferon beta-l a), BETASERON® (Berlex) (Interferon beta- Ib), COPAXONE® (Teva Neuroscience) (glatiramer acetate injection) and REB1F® (Pfizer) (interferon beta- 1 a). In addition, macrolide and/or mycophenolic acid, which has multiple activities, can be co-administered with a formulation of the invention. Macrolide antibiotics include tacrolimus, cyclosporine, sirolimus, everolimus, ascomycin, erythromycin, azithromycin, clarithromycin, clindamycin, lincomycin, dirithromycin, josamycin, spiramycin, diacetyl-midecamycin, tylosin, roxithromycin, ABT-773, telithromycin, leucomycins, and lincosamide. Mitochondrial-Associated Diseases and Disorders In certain embodiments, the invention provides methods for treating diseases or disorders that would benefit from increased mitochondrial activity. The methods involve administering to a subject in need thereof a therapeutically effective amount of a formulation of the invention. Increased mitochondrial activity refers to increasing activity of the mitochondria while maintaining the overall numbers of mitochondria (e.g., mitochondrial mass), increasing the numbers of mitochondria thereby increasing mitochondrial activity (e.g., by stimulating mitochondrial biogenesis), or combinations thereof. In certain embodiments, diseases and disorders that would benefit from increased mitochondrial activity include diseases or disorders associated with mitochondrial dysfunction. In certain embodiments, methods for treating diseases or disorders that would benefit from increased mitochondrial activity may comprise identifying a subject suffering from a mitochondrial dysfunction. Methods for diagnosing a mitochondrial dysfunction may involve molecular genetic, pathologic and/or biochemical analysis are summarized in Cohen and Gold, Cleveland Clinic Journal- of Medicine, 68: 625-642 (2001 ). One method for diagnosing a mitochondrial dysfunction is the Thor-Byrne-ier scale (see e.g., Cohen and Gold, supra; Collin S. et al., Eur Neurol. 36: 260-267 (1996)). Other methods for determining mitochondrial number and function include, for example, enzymatic assays (e.g., a mitochondrial enzyme or an ATP biosynthesis factor such as an ETC enzyme or a Krebs cycle enzyme), determination or mitochondrial mass, mitochondrial volume, and/or mitochondrial number, quantification of mitochondrial DNA, monitoring intracellular calcium homeostasis and/or cellular responses to perturbations of this homeostasis, evaluation of response to an apoptogenic stimulus, determination of free radical production. Such methods are known in the art and are described, for example, in U.S. Patent Publication No. 2002/0049176 and references cited therein.
Mitochondria are critical for the survival and proper function of almost all types of eukaryotic cells. Mitochondria in virtually any cell type can have congenital or acquired defects that affect their function. Thus, the clinically significant signs and symptoms of mitochondrial defects affecting respiratory chain function are heterogeneous and variable depending on the distribution of defective mitochondria among cells and the severity of their deficits, and upon physiological demands upon the affected cells. Nondividing tissues with high energy requirements, e.g. nervous tissue, skeletal muscle and cardiac muscle are particularly susceptible to mitochondrial respiratory chain dysfunction, but any organ system can be affected.
Diseases and disorders associated with mitochondrial dysfunction include diseases and disorders in which deficits in mitochondrial respiratory chain activity contribute to the development of pathophysiology of such diseases or disorders in a mammal. This includes 1) congenital genetic deficiencies in activity of one or more components of the mitochondrial respiratory chain; and 2) acquired deficiencies in the activity of one or more components of the mitochondrial respiratory chain, wherein such deficiencies are caused by a) oxidative damage during aging; b) elevated intracellular calcium; c) exposure of affected cells to nitric oxide; d) hypoxia or ischemia; e) microtubule-associated deficits in axonal transport of mitochondria, or f) expression of mitochondrial uncoupling proteins.
Diseases or disorders that would benefit from increased mitochondrial activity generally include for example, diseases in which free radical mediated oxidative injury leads to tissue degeneration, diseases in which cells inappropriately undergo apoptosis, and diseases in which cells fail to undergo apoptosis. Exemplary diseases or disorders that would benefit from increased mitochondrial activity include, for example, AD (Alzheimer's Disease), ADPD (Alzheimer's Disease and Parkinsons's Disease), AMDF (Ataxia, Myoclonus and Deafness), auto-immune disease, cancer, CIPO (Chronic Intestinal Pseudoobstruction with myopathy and Ophthalmoplegia), congenital muscular dystrophy, CPEO (Chronic Progressive External Ophthalmoplegia), DEAF (Maternally inherited DEAFness or aminoglycoside-induced DEAFness), DEMCHO (Dementia and Chorea), diabetes mellitus (Type I or Type II), DIDMOAD (Diabetes Insipidus, Diabetes Mellitus, Optic Atrophy, Deafness), DMDF (Diabetes Mellitus and Deafness), dystonia, Exercise Intolerance, ESOC (Epilepsy, Strokes, Optic atrophy, and Cognitive decline), FBSN (Familial Bilateral Striatal Necrosis), FICP (Fatal Infantile Cardiomyopathy Plus, a MELAS-associated cardiomyopathy), GER (Gastrointestinal Reflux), HD (Huntington's Disease), KSS (Kearns Sayre Syndrome), "later-onset" myopathy, LDYT (Leber's hereditary optic neuropathy and DYsTonia), Leigh's Syndrome, LHON (Leber Hereditary Optic Neuropathy), LIMM (Lethal Infantile Mitochondrial Myopathy), MDM (Myopathy and Diabetes Mellitus), MELAS (Mitochondrial Encephalomyopathy, Lactic Acidosis, and Stroke-like episodes), MEPR (Myoclonic Epilepsy and Psychomotor Regression), MERME (MERRF/MELAS overlap disease), MERRF (Myoclonic Epilepsy and Ragged Red Muscle Fibers), MHCM (Maternally Inherited Hypertrophic CardioMyopathy), MICM (Maternally Inherited Cardiomyopathy), MILS
(Maternally Inherited Leigh Syndrome), Mitochondrial Encephalocardiomyopafhy, Mitochondrial Encephalomyopathy, MM (Mitochondrial Myopathy), MMC (Maternal Myopathy and Cardiomyopathy), MNGIE (Myopathy and external ophthalmoplegia, Neuropathy, Gastro-Intestinal, Encephalopathy), Multisystem Mitochondrial Disorder (myopathy, encephalopathy, blindness, hearing loss, peripheral neuropathy), NARP (Neurogenic muscle weakness, Ataxia, and Retinitis Pigmentosa; alternate phenotype at this locus is reported as Leigh Disease), PD (Parkinson's Disease), Pearson's Syndrome, PEM (Progressive Encephalopathy), PEO (Progressive External Ophthalmoplegia), PME (Progressive Myoclonus Epilepsy), PMPS (Pearson Marrow-Pancreas Syndrome), psoriasis, RTT (Rett Syndrome), schizophrenia, SIDS (Sudden Infant Death Syndrome), SNHL (Sensorineural Hearing Loss), Varied Familial Presentation (clinical manifestations range from spastic paraparesis to multisystem progressive disorder & fatal cardiomyopathy to truncal ataxia, dysarthria, severe hearing loss, mental regression, ptosis, ophthalmoparesis, distal cyclones, and diabetes mellitus), or Wolfram syndrome.
. Other diseases and disorders that would benefit from increased mitochondrial activity include, for example, Friedreich's ataxia and other ataxias, amyotrophic lateral sclerosis (ALS) and other motor neuron diseases, macular degeneration, epilepsy, Alpers syndrome, Multiple mitochondrial DNA deletion syndrome, MtDNA depletion syndrome, Complex I deficiency, Complex II (SDH) deficiency, Complex III deficiency, Cytochrome c oxidase (COX, Complex IV) deficiency, Complex V deficiency, Adenine Nucleotide Translocator (ANT) deficiency, Pyruvate dehydrogenase (PDH) deficiency, Ethylmalonic aciduria with lactic acidemia, 3-Methyl glutaconic aciduria with lactic acidemia, Refractory epilepsy with declines during infection, Asperger syndrome with declines during infection, Autism with declines during infection, Attention deficit hyperactivity disorder (ADHD), Cerebral palsy with declines during infection, Dyslexia with declines during infection, materially inherited thrombocytopenia and leukemia syndrome, MARIAHS syndrome (Mitrochondrial ataxia, recurrent infections, aphasia, hypouricemia/hypomyelination, seizures, and dicarboxylic aciduria), ND6 dystonia, Cyclic vomiting syndrome with declines during infection, 3-Hydroxy isobutryic aciduria with lactic acidemia, Diabetes mellitus with lactic acidemia, Uridine responsive neurologic syndrome (URNS), Dilated cardiomyopathy, Splenic Lymphoma, and Renal Tubular Acidosis/Diabetes/Ataxis syndrome.
In other embodiments, the invention provides methods for treating a subject suffering from mitochondrial disorders arising from, but not limited to, posttraumatic head injury and cerebral edema, stroke (invention methods useful for preventing or preventing reperfusion injury), Lewy body dementia, hepatorenal syndrome, acute liver failure, NASH (non-alcoholic steatohepatitis), Anti- metastasis/prodifferentiation therapy of cancer, idiopathic congestive heart failure, atrial fibrilation (non-valvular), Wolff-Parkinson- White Syndrome, idiopathic heart block, prevention of reperfusion injury in acute myocardial infarctions, familial migraines, irritable bowel syndrome, secondary prevention of non-Q wave myocardial infarctions, Premenstrual syndrome, Prevention of renal failure in hepatorenal syndrome, anti-phospholipid antibody syndrome, eclampsia/pre- eclampsia, oopause infertility, ischemic heart disease/angina, and Shy-Drager and unclassified dysautonomia syndromes.
In still another embodiment, there are provided methods for the treatment of mitochondrial disorders associated with pharmacological drug-related side effects. Types of pharmaceutical agents that are associated with mitochondrial disorders include reverse transcriptase inhibitors, protease inhibitors, inhibitors of DHOD, and the like. Examples of reverse transcriptase inhibitors include, for example, Azidothymidine (AZT), Stavudine (D4T), Zalcitabine (ddC), Didanosine (DDI), Fluoroiodoarauracil (FIAU), Lamivudine (3TC), Abacavir and the like. Examples of protease inhibitors include, for example, Ritonavir, Indinavir, Saquinavir, Nelfinavir and the like. Examples of inhibitors of dihydroorotate dehydrogenase (DHOD) include, for example, Leflunomide, Brequinar, and the like. Reverse transcriptase inhibitors not only inhibit reverse transcriptase but also polymerase gamma which is required for mitochondrial function. Inhibition of polymerase gamma activity (e.g., with a reverse transcriptase inhibitor) therefore leads to mitochondrial dysfunction and/or a reduced mitochondrial mass which manifests itself in patients as hyperlactatemia. This type of condition may benefit from an increase in the number of mitochondria and/or an improvement in mitochondrial function, e.g., by administration of a formulation of the invention.
Common symptoms of mitochondrial diseases include cardiomyopathy, muscle weakness and atrophy, developmental delays (involving motor, language, cognitive or executive function), ataxia, epilepsy, renal tubular acidosis, peripheral neuropathy, optic neuropathy, autonomic neuropathy, neurogenic bowel dysfunction, sensorineural deafness, neurogenic bladder dysfunction, dilating cardiomyopathy, migraine, hepatic failure, lactic acidemia, and diabetes mellitus.
In certain embodiments, the invention provides methods for treating a disease or disorder that would benefit from increased mitochondrial activity that involves administering to a subject in need thereof one or more sirtuin activating compounds in combination with another therapeutic agent such as, for example, an agent useful for treating mitochondrial dysfunction (such as antioxidants, vitamins, or respiratory chain cofactors), an agent useful for reducing a symptom associated with a disease or disorder involving mitochondrial dysfunction (such as, an anti- seizure agent, an agent useful for alleviating neuropathic pain, an agent for treating cardiac dysfunction), a cardiovascular agent (as described further below), a chemotherapeutic agent (as described further below), or an anti-neurodegeneration agent (as described further below). In an exemplary embodiment, the invention provides methods for treating a disease or disorder that would benefit from increased mitochondrial activity that involves administering to a subject in need thereof one or more sirtuin activating compounds in combination with one or more of the following: coenzyme Qjo, L-carnitine, thiamine, riboflavin, niacinamide, folate, vitamin E, selenium, lipoic acid, or prednisone. Compositions comprising such combinations are also provided herein.
In exemplary embodiments, the invention provides methods for treating diseases or disorders that would benefit from increased mitochondrial acitivty by administering to a subject a therapeutically effective amount of a formulation of the invention. Exemplary diseases or disorders include, for example, neuromuscular disorders (e.g., Friedreich's Ataxia, muscular dystrophy, multiple sclerosis, etc.), disorders of neuronal instability (e.g., seizure disorders, migrane, etc.), developmental delay, neurodegenerative disorders (e.g., Alzheimer's Disease, Parkinson's Disease, amyotrophic lateral sclerosis, etc.), ischemia, renal tubular acidosis, age-related neurodegeneration and cognitive decline, chemotherapy fatigue, age-related or chemotherapy-induced menopause or irregularities of menstrual cycling or ovulation, mitochondrial myopathies, mitochondrial damage (e.g., calcium accumulation, excitotoxicity, nitric oxide exposure, hypoxia, etc.), and mitochondrial deregulation.
A gene defect underlying Friedreich's Ataxia (FA), the most common hereditary ataxia, was recently identified and is designated "frataxin". In FA, after a period of normal development, deficits in coordination develop which progress to paralysis and death, typically between the ages of 30 and 40. The tissues affected - most severely are the spinal cord, peripheral nerves, myocardium, and pancreas. Patients typically lose motor control and are confined to wheel chairs, and are commonly afflicted with heart failure and diabetes. The genetic basis for FA involves GAA trinucleotide repeats in an intron region of the gene encoding frataxin. The presence of these repeats results in reduced transcription and expression of the gene. Frataxin is involved in regulation of mitochondrial iron content. When cellular frataxin content is subnormal, excess iron accumulates in mitochondria, promoting oxidative damage and consequent mitochondrial degeneration and dysfunction. When intermediate numbers of GAA repeats are present in the frataxin gene intron, the severe clinical phenotype of ataxia may not develop. However, these intermediate-length trinucleotide extensions are found in 25 to 30% of patients with non-insulin dependent diabetes mellitus, compared to about 5% of the nondiabetic population. In certain embodiments, sirtuin activating compounds may be used for treating patients with disorders related to deficiencies or defects in frataxin, including Friedreich's Ataxia, myocardial dysfunction, diabetes mellitus and complications of diabetes like peripheral neuropathy.
Muscular dystrophy refers to a family of diseases involving deterioration of neuromuscular structure and function, often resulting in atrophy of skeletal muscle and myocardial dysfunction. In the case of Duchenne muscular dystrophy, mutations or deficits in a specific protein, dystrophin, are implicated in its etiology. Mice with their dystrophin genes inactivated display some characteristics of muscular dystrophy, and have an approximately 50% deficit in mitochondrial respiratory chain activity. A final common pathway for neuromuscular degeneration in most cases is calcium-mediated impairment of mitochondrial function. In certain embodiments, sirtuin activating compounds may be used for reducing the rate of decline in muscular functional capacities and for improving muscular functional status in patients with muscular dystrophy. Multiple sclerosis (MS) is a neuromuscular disease characterized by focal inflammatory and autoimmune degeneration of cerebral white matter. Periodic exacerbations or attacks are significantly correlated with upper respiratory tract and other infections, both bacterial and viral, indicating that mitochondrial dysfunction plays a role in MS. Depression of neuronal mitochondrial respiratory chain activity caused by Nitric Oxide (produced by astrocytes and other cells involved in inflammation) is implicated as a molecular mechanism contributing to MS. In certain embodiments, sirtuin activating compounds may be used for treatment of patients with multiple sclerosis, both prophylactically and during episodes of disease exacerbation. Epilepsy is often present in patients with mitochondrial cytopathies, involving a range of seizure severity and frequency, e.g. absence, tonic, atonic, myoclonic, and status epilepticus, occurring in isolated episodes or many times daily. In certain embodiments, sirtuin activating compounds may be used for treating patients with seizures secondary to mitochondrial dysfunction, including reducing frequency and severity of seizure activity.
Metabolic studies on patients with recurrent migraine headaches indicate that deficits in mitochondrial activity are commonly associated with this disorder, manifesting as impaired-oxidative phosphorylation and excess lactate production. Such deficits are not necessarily due to genetic defects in mitochondrial DNA. Migraineurs are hypersensitive to nitric oxide, an endogenous inhibitor of Cytochrome c Oxidase. In addition, patients with mitochondrial cytopathies, e.g. MELAS, often have recurrent migraines. In certain embodiments, sirtuin activating compounds may be used for treating patients with recurrent migraine headaches, including headaches refractory to ergot compounds or serotonin receptor antagonists.
Delays in neurological or neuropsychological development are often found in children with mitochondrial diseases. Development and remodeling of neural connections requires intensive biosynthetic activity, particularly involving synthesis of neuronal membranes and myelin, both of which require pyrimidine nucleotides as cofactors. Undine nucleotides are involved inactivation and transfer of sugars to glycolipids and glycoproteins. Cytidine nucleotides are derived from uridine nucleotides, and are crucial for synthesis of major membrane phospholipid constituents like phosphatidylcholine, which receives its choline moiety from cytidine diphosphocholine. In the case of mitochondrial dysfunction (due to either mitochondrial DNA defects or any of the acquired or conditional deficits like exicitoxic or nitric oxide-mediated mitochondrial dysfunction) or other conditions resulting in impaired pyrimidine synthesis, cell proliferation and axonal extension is impaired at crucial stages in development of neuronal interconnections and circuits, resulting in delayed or arrested development of neuropsychological functions like language, motor, social, executive function, and cognitive skills. In autism for example, magnetic resonance spectroscopy measurements of cerebral phosphate compounds indicates that there is global undersynthesis of membranes and membrane. precursors indicated by reduced levels of uridine diphospho-sugars, and cytidine nucleotide derivatives involved in membrane synthesis. Disorders characterized by developmental delay include Rett's Syndrome, pervasive developmental delay (or PDD-NOS "pervasive developmental delay not otherwise specified" to distinguish it from specific subcategories like autism), autism,
Asperger's Syndrome, and Attention Deficit/Hyperactivity Disorder (ADHD), which is becoming recognized as a delay or lag in development of neural circuitry underlying executive functions. In certain embodiments, sirtuin activating compounds may be useful for treating treating patients with neurodevelopmental delays (e.g., involving motor, language, executive function, and cognitive skills), or other delays or arrests of neurological and neuropsychological development in the nervous system and somatic development in non-neural tissues like muscle and endocrine glands.
The two most significant severe neurodegenerative diseases associated with aging, Alzheimer's Disease (AD) and Parkinson's Disease (PD), both involve mitochondrial dysfunction in their pathogenesis. Complex 1 deficiencies in particular are frequently found not only in the nigrostriatal neurons that degenerate in
Parkinson's disease, but also in peripheral tissues and cells like muscle and platelets of Parkinson's Disease patients. In Alzheimer's Disease, mitochondrial respiratory chain activity is often depressed, especially Complex IV (Cytochrome c Oxidase). Moreover, mitochondrial respiratory function altogether is depressed as a consequence of aging, further amplifying the deleterious sequelae of additional molecular lesions affecting respiratory chain function. Other factors in addition to primary mitochondrial dysfunction underlie neurodegeneration in AD, PD, and related disorders. Excitotoxic stimulation and nitric oxide are implicated in both diseases, factors which both exacerbate mitochondrial respiratory chain deficits and whose deleterious actions are exaggerated on a background of respiratory chain dysfunction. Huntington's Disease also involves mitochondrial dysfunction in affected brain regions, with cooperative interactions of excitotoxic stimulation and mitochondrial dysfunction contributing to neuronal degeneration. In certain embodiments, sirtuin activating compounds may be useful for treating and attenuating progression of age-related neurodegenerative diseases including AD and PD.
One of the major genetic defects in patients with Amyotrophic Lateral Sclerosis (ALS or Lou Gehrig's Disease) is mutation or deficiency in Copper-Zinc Superoxide Dismutase (SOD 1 ), an antioxidant enzyme. Mitochondria both produce and are primary targets for reactive oxygen species. Inefficient transfer of electrons to oxygen in mitochondria is the most significant physiological source of free radicals in mammalian systems. Deficiencies in antioxidants or antioxidant enzymes can result in or exacerbate mitochondrial degeneration. Mice transgenic for mutated SODl develop symptoms and pathology similar to those in human ALS. The development of the disease in these animals has been shown to involve oxidative destruction of mitochondria followed by functional decline of motor neurons and onset of clinical symptoms. Skeletal muscle from ALS patients has low mitochondrial Complex I activity. In certain embodiments, sirtuin activating compounds may be useful for treating ALS, for reversing or slowing the progression of clinical symptoms.
Oxygen deficiency results in both direct inhibition of mitochondrial respiratory chain activity by depriving cells of a terminal electron acceptor for
Cytochrome c reoxidation at Complex IV, and indirectly, especially in the nervous system, via secondary post-anoxic excitotoxicity and nitric oxide formation. In conditions like cerebral anoxia, angina or sickle cell anemia crises, tissues are relatively hypoxic. In such cases, compounds that increase mitochondrial activity provide protection of affected tissues from deleterious effects of hypoxia, attenuate secondary delayed cell death, and accelerate recovery from hypoxic tissue stress and injury. In certain embodiments, sirtuin activating compounds may be useful for preventing delayed cell death (apoptosis in regions like the hippocampus or cortex occurring about 2 to 5 days after an episode of cerebral ischemia) after ischemic or hypoxic insult to the brain.
Acidosis due to renal dysfunction is often observed in patients with mitochondrial disease, whether the underlying respiratory chain dysfunction is congenital or induced by ischemia or cytotoxic agents like cisplatin. Renal tubular acidosis often requires administration of exogenous sodium bicarbonate to maintain blood and tissue pH. In certain embodiments, sirtuin activating compounds may be useful for treating renal tubular acidosis and other forms of renal dysfunction caused by mitochondrial respiratory chain deficits.
During normal aging, there is a progressive decline in mitochondrial respiratory chain function. Beginning about age 40, there is an exponential rise in accumulation of mitochondrial DNA defects in humans, and a concurrent decline in nuclear-regulated elements of mitochondrial respiratory activity. Many mitochondrial DNA lesions have a selection advantage during mitochondrial turnover, especially in postmitotic cells. The proposed mechanism is that mitochondria with a defective respiratory chain produce less oxidative damage to themselves than do mitochondria with intact functional respiratory chains (mitochondrial respiration is the primary source of free radicals in the body). Therefore, normally-functioning mitochondria accumulate oxidative damage to membrane lipids more rapidly than do defective mitochondria, and are therefore "tagged" for degradation by lysosomes. Since mitochondria within cells have a half life of about 10 days, a selection advantage can result in rapid replacement of functional mitochondria with those with diminished respiratory activity, especially in slowly dividing cells. The net result is that once a mutation in a gene for a mitochondrial protein that reduces oxidative damage to mitochondria occurs, such defective mitochondria, will rapidly populate the cell, diminishing or eliminating its respiratory capabilities. The accumulation of such cells results in aging or degenerative disease at the organismal level. This is consistent with the progressive mosaic appearance of cells with defective electron transport activity in muscle, with cells almost devoid of Cytochrome c Oxidase (COX) activity interspersed randomly amidst cells with normal activity, and a higher incidence of COX-negative cells in biopsies from older subjects. The organism, during aging, or in a variety of mitochondrial diseases, is thus faced with a situation in which irreplaceable postmitotic cells (e.g. neurons, skeletal and cardiac muscle) must be preserved and their function maintained to a significant degree, in the face of an inexorable progressive decline in mitochondrial respiratory chain function. Neurons with dysfunctional mitochondria become progressively more sensitive to insults like excitotoxic injury. Mitochondrial failure contributes to most degenerative diseases (especially neurodegeneration) that accompany aging. Congenital mitochondrial diseases often involve early-onset neurodegeneration similar in fundamental mechanism to disorders that occur during aging of people born with normal mitochondria. In certain embodiments, sirtuin activating compounds may be useful for treating or attenuating cognitive decline and other degenerative consequences of aging.
Mitochondrial DNA damage is more extensive and persists longer than nuclear DNA damage in cells subjected to oxidative stress or cancer chemotherapy agents like cisplatin due to both greater vulnerability and less efficient repair of mitochondrial DNA. Although mitochondrial DNA may be more sensitive to damage than nuclear DNA, it is relatively resistant, in some situations, to mutagenesis by chemical carcinogens. This is because mitochondria respond to some types of mitochondrial DNA damage by destroying their defective genomes rather than attempting to repair them. This results in global mitochondrial dysfunction for a period after cytotoxic chemotherapy. Clinical use of chemotherapy agents like cisplatin, mitomycin, and Cytoxan is often accompanied by debilitating "chemotherapy fatigue", prolonged periods of weakness and exercise intolerance which may persist even after recovery from hematologic and gastrointestinal toxicities of such agents. In certain embodiments, formulations of the invention may be useful for treatment and prevention of side effects of cancer chemotherapy related to mitochondrial dysfunction.
A crucial function of the ovary is to maintain integrity of the mitochondrial genome in oocytes, since mitochondria passed onto a fetus are all derived from those present in oocytes at the time of conception. Deletions in mitochondrial DNA become detectable around the age of menopause, and are also associated with abnormal menstrual cycles. Since cells cannot directly detect and respond to defects in mitochondrial DNA, but can only detect secondary effects that affect the cytoplasm, like impaired respiration, redox status, or deficits in pyrimidine synthesis, such products of mitochondrial function participate as a signal for oocyte selection and follicular atresia, ultimately triggering menopause when maintenance of mitochondrial genomic fidelity and functional activity can no longer be guaranteed. This is analogous to apoptosis in cells with DNA damage, which undergo an active process of cellular suicide when genomic fidelity can no longer be achieved by repair processes. Women with mitochondrial cytopathies affecting the gonads often undergo premature menopause or display primary cycling abnormalities. Cytotoxic cancer chemotherapy often induces premature menopause, with a consequent increased risk of osteoporosis. Chemotherapy-induced amenorrhea is generally due to primary ovarian failure. The incidence of chemotherapy-induced amenorrhea increases as a function of age in premenopausal women receiving chemotherapy, pointing toward mitochondrial involvement. Inhibitors of mitochondrial respiration or protein synthesis inhibit hormone-induced ovulation, and furthermore inhibit production of ovarian steroid hormones in response to pituitary gonadotropins. Women with Down's syndrome typically undergo menopause prematurely, and also are subject to early onset of Alzheimer- like dementia. Low activity of cytochrome oxidase is consistently found in tissues of Down's patients and in late-onset Alzheimer's Disease. Appropriate support of mitochondrial function or compensation for mitochondrial dysfunction therefore is useful for protecting against age-related or chemotherapy-induced menopause or irregularities of menstrual cycling or ovulation. In certain embodiments, formulations of the invention may be useful for treating and preventing amenorrhea, irregular ovulation, menopause, or secondary consequences of menopause.
In certain embodiments, formulations of the invention may be useful for treatment mitochondrial myopathies. Mitochondrial myopathies range from mild, slowly progressive weakness of the extraocular muscles to severe, fatal infantile myopathies and multisystem encephalomyopathies. Some syndromes have been defined, with some overlap between them. Established syndromes affecting muscle include progressive external ophthalmoplegia, the Kearns-Sayre syndrome (with ophthalmoplegia, pigmentary retinopathy, cardiac conduction defects, cerebellar ataxia, and sensorineural deafness), the MELAS syndrome (mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes), the MERFF syndrome (myoclonic epilepsy and ragged red fibers), limb-girdle distribution weakness, and infantile myopathy (benign or severe and fatal). Muscle biopsy specimens stained with modified Gomori's trichrome stain show ragged red fibers due to excessive accumulation of mitochondria. Biochemical defects in substrate transport and utilization, the Krebs cycle, oxidative phosphorylation, or the respiratory chain are detectable. Numerous mitochondrial DNA point mutations and deletions have been described, transmitted in a maternal, nonmendelian inheritance pattern. Mutations in nuclear-encoded mitochondrial enzymes occur.
In certain embodiments, formulations of the invention may be useful for treating patients suffering from toxic damage to mitochondria, such as, toxic damage due to calcium accumulation, excitotoxicity, nitric oxide exposure, drug induced toxic damage, or hypoxia. A fundamental mechanism of cell injury, especially in excitable tissues, involves excessive calcium entry into cells, as a result of either leakage through the plasma membrane or defects in intracellular calcium handling mechanisms. Mitochondria are major sites of calcium sequestration, and preferentially utilize energy from the respiratory chain for taking up calcium rather than for ATP synthesis, which results in a downward spiral of mitochondrial failure, since calcium uptake into mitochondria results in diminished capabilities for energy transduction.
Excessive stimulation of neurons with excitatory amino acids is a common mechanism of cell death or injury in the central nervous system. Activation of glutamate receptors, especially of the subtype designated NMDA receptors, results in mitochondrial dysfunction, in part through elevation of intracellular calcium during excitotoxic stimulation. Conversely, deficits in mitochondrial respiration and oxidative phosphorylation sensitizes cells to excitotoxic stimuli, resulting in cell death or injury during exposure to levels of excitotoxic neurotransmitters or toxins that would be innocuous to normal cells.
Nitric oxide (about 1 micromolar) inhibits cytochrome oxidase (Complex IV) and thereby inhibits mitochondrial respiration; moreover, prolonged exposure to nitric oxide (NO) irreversibly reduces Complex 1 activity. Physiological or pathophysiological concentrations of NO thereby inhibit pyrimidine biosynthesis. Nitric oxide is implicated in a variety of neurodegenerative disorders including inflammatory and autoimmune diseases of the central nervous system, and is involved in mediation of excitotoxic and post-hypoxic damage to neurons.
Oxygen is the terminal electron acceptor in the respiratory chain. Oxygen deficiency impairs electron transport chain activity, resulting in diminished pyrimidine synthesis as well as diminished ATP synthesis via oxidative phosphorylation. Human cells proliferate and retain viability under virtually anaerobic conditions if provided with uridine and pyruvate (or a similarly effective agent for oxidizing NADH to optimize glycolytic ATP production).
In certain embodiments, formulations of the invention may be useful for treating diseases or disorders associated with mitochondrial deregulation.
Transcription of mitochondrial DNA encoding respiratory chain components requires nuclear factors. In neuronal axons, mitochondria must shuttle back and forth to the nucleus in order to maintain respiratory chain activity. If axonal transport is impaired by hypoxia or by drugs like taxol which affect microtubule stability, mitochondria distant from the nucleus undergo loss of cytochrome oxidase activity. Accordingly, treatment with a formulation of the invention may be useful for promoting nuclear-mitochondrial interactions.
Mitochondria are the primary source of free radicals and reactive oxygen species, due to spillover from the mitochondrial respiratory chain, especially when defects in one or more respiratory chain components impairs orderly transfer of electrons from metabolic intermediates to molecular oxygen. To reduce oxidative damage, cells can compensate by expressing mitochondrial uncoupling proteins (UCP), of which several have been identified. UCP-2 is transcribed in response to oxidative damage, inflammatory cytokines, or excess lipid loads, e.g. fatty liver and steatohepatitis. UCPs reduce spillover of reactive oxygen species from mitochondria by discharging proton gradients across the mitochondrial inner membrane, in effect wasting energy produced by metabolism and rendering cells vulnerable to energy stress as a trade-off for reduced oxidative injury. Muscle Performance
In other embodiments, the invention provides methods for enhancing muscle performance by administering a therapeutically effective amount of a formulation of the invention. For example, formulations of the invention may be useful for improving physical endurance (e.g., ability to perform a physical task such as exercise, physical labor, sports activities, etc.), inhibiting or retarding physical fatigues, enhancing blood oxygen levels, enhancing energy in healthy individuals, enhance working capacity and endurance, reducing muscle fatigue, reducing stress, enhancing cardiac and cardiovascular function, improving sexual ability, increasing muscle ATP levels, and/or reducing lactic acid in blood. In certain embodiments, the methods involve administering an amount of a formulation of the invention that increase mitochondrial activity, increase mitochondrial biogenesis, and/or increase mitochondrial mass. Sports performance refers to the ability of the athlete's muscles to perform when participating in sports activities. Enhanced sports performance, strength, speed and endurance are measured by an increase in muscular contraction strength, increase in amplitude of muscle contraction, shortening of muscle reaction time between stimulation and contraction. Athlete refers to an individual who participates in sports at any level and who seeks to achieve an improved level of strength, speed and endurance in their performance, such as, for example, body builders, bicyclists, long distance runners, short distance runners, etc. An athlete may be hard training, that is, performs sports activities intensely more than three days a week or for competition. An athlete may also be a fitness enthusiast who seeks to improve general health and well-being, improve energy levels, who works out for about 1-2 hours about 3 times a week. Enhanced sports performance in manifested by the ability to overcome muscle fatigue, ability to maintain activity for longer periods of time, and have a more effective workout.
In the arena of athlete muscle performance, it is desirable to create conditions that permit competition or training at higher levels of resistance for a prolonged period of time. However, acute and intense anaerobic use of skeletal muscles often results in impaired athletic performance, with losses in force and work output, and increased onset of muscle fatigue, soreness, and dysfunction. It is now recognized that even a single exhaustive exercise session, or for that matter any acute trauma to the body such as muscle injury, resistance or exhaustive muscle exercise, or elective surgery, is characterized by perturbed metabolism that affects muscle performance in both short and long term phases. Both muscle metabolic/enzymatic activity and gene expression are affected. For example, disruption of skeletal muscle nitrogen metabolism as well as depletion of sources of metabolic energy occur during extensive muscle activity. Amino acids, including branched-chain amino acids, are released from muscles followed by their deamination to elevate serum ammonia and local oxidation as muscle fuel sources, which augments metabolic acidosis. In addition, there is a decline in catalytic efficiency of muscle contraction events, as well as an alteration of enzymatic activities of nitrogen and energy metabolism. Further, protein catabolism is initiated where rate of protein synthesis is decreased coupled with an increase in the degradation of non-contractible protein. These metabolic processes are also accompanied by free radical generation which further damages muscle cells. Recovery from fatigue during acute and extended exercise requires reversal of metabolic and non-metabolic fatiguing factors. Known factors that participate in human muscle fatigue, such as lactate, ammonia, hydrogen ion, etc., provide an incomplete and unsatisfactory explanation of the fatigue/recovery process, and it is likely that additional unknown agents participate (Baker et al., J. Appl. Physiol. 74:2294-2300, 1993; Bazzarre et al., J Am. Coll. Nutr. 1 1 :505-51 1, 1992; Dohm et al., Fed. Proc. 44:348-352, 1985; Edwards In: Biochemistry of Exercise, Proceedings of the Fifth International Symposium on the Biochemistry of Exercise (Kutrgen, Vogel, Poormans, eds.), 1983; MacDougall et al., Acta Physiol. Scand. 146:403-404, 1992; Walser et al., Kidney Int. 32: 123-128, 1987). Several studies have also analyzed the effects of nutritional supplements and herbal supplements in enhancing muscle performance.
Aside from muscle performance during endurance exercise, free radicals and oxidative stress parameters are affected in pathophysiological states. A substantial body of data now suggests that oxidative stress contributes to muscle wasting or atrophy in pathophysiological states (reviewed in Clarkson, P. M. Antioxidants and physical performance. Crit. Rev. Food Sci. Nutr. 35: 31 -41 ; 1995; Powers, S. K.; Lennon, S. L. Analysis of cellular responses to free radicals: Focus on exercise and skeletal muscle. Proc. Nutr. Soc. 58: 1025-1033; 1999). For example, with respect to muscular disorders where both muscle endurance and function are compensated, the role of nitric oxide (NO), has been implicated. In muscular dystrophies, especially those due to defects in proteins that make up the dystrophin-glycoprotein complex (DGC), the enzyme that synthesizes NO, nitric oxide synthase (NOS), has been associated. Recent studies of dystrophies related to DGC defects suggest that one mechanism of cellular injury is functional ischemia related to alterations in cellular NOS and disruption of a normal protective action of NO. This protective action is the prevention of local ischemia during contraction-induced increases in sympathetic vasoconstriction. Rando (Microsc Res Tech 55(4):223-35, 2001 ), has shown that oxidative injury precedes pathologic changes and that muscle cells with defects in the DGC have an increased susceptibility to oxidant challenges. Excessive lipid peroxidation due to free radicals has also been shown to be a factor in myopathic diseases such as McArdle's disease (Russo et al., Med Hypotheses. 39(2): 147-51 , 1992). Furthermore, mitochondrial dysfunction is a well-known correlate of age- related muscle wasting (sarcopenia) and free radical damage has been suggested, though poorly investigated, as a contributing factor (reviewed in Navarro, A.; Lopez-Cepero, J. M.; Sanchez del Pino, M. L. Front. Biosci. 6: D26-44; 2001). Other indications include acute sarcopenia, for example muscle atrophy and/or cachexia associated with burns, bed rest, limb immobilization, or major thoracic, abdominal, and/or orthopedic surgery. It is contemplated that the methods of the present invention will also be effective in the treatment of muscle related pathological conditions. In certain embodiments, the invention provides novel dietary compositions comprising formulations of the invention, a method for their preparation, and a method of using the compositions for improvement of sports performance. Accordingly, provided are therapeutic compositions, foods and beverages that have actions of improving physical endurance and/or inhibiting physical fatigues for those people involved in broadly-defined exercises including sports requiring endurance and labors requiring repeated muscle exertions. Such dietary compositions may additional comprise electrolytes, caffeine, vitamins, carbohydrates, etc. Other Uses
Formulations of the invention may be used for treating orpreventing viral infections (such as infections by influenza, herpes or papilloma virus) or as antifungal agents. In certain embodiments, formulations of the invention may be administered as part of a combination drug therapy with another therapeutic agent for the treatment of viral diseases, including, for example, acyclovir, ganciclovir and zidovudine. In another embodiment, formulations of the invention may be administered as part of a combination drug therapy with another anti-fungal agent including, for example, topical anti-fungals such as ciclopirox, clotrimazole, econazole, miconazole, nystatin, oxiconazole, terconazole, and tolnaftate, or systemic anti-fungal such as fluconazole (Diflucan), itraconazole (Sporanox), ketoconazole (Nizoral), and miconazole (Monistat I.V.). Subjects that may be treated as described herein include eukaryotes, such as mammals, e.g., humans, ovines, bovines, equines, porcines, canines, felines, non- human primate, mice, and rats. Cells that may be treated include eukaryotic cells, " e.g., from a subject described above, or plant cells, yeast cells and prokaryotic cells, e.g., bacterial cells. For example, formulations of the invention may be administered to farm animals to improve their ability to withstand farming conditions longer. Formulations of the invention may also be used to increase lifespan, stress resistance, and resistance to apoptosis in plants. In one embodiment, a formulation of the invention is applied to plants, e.g., on a periodic basis, or to fungi. In another embodiment, plants are genetically modified to produce a compound. In another embodiment, plants and fruits are treated with a formulation of the invention prior to picking and shipping to increase resistance to damage during shipping. Plant seeds may also be contacted with a formulation of the invention to preserve them.
In other embodiments, formulations of the invention may be used for modulating lifespan in yeast cells. Situations in which it may be desirable to extend the lifespan of yeast cells include any process in which yeast is used, e.g., the making of beer, yogurt, and bakery items, e.g., bread. Use of yeast having an extended lifespan can result in using less yeast or in having the yeast be active for longer periods of time. Yeast or other mammalian cells used for recombinantly producing proteins may also be treated as described herein.
Formulations of the invention may also be used to increase lifespan, stress resistance and resistance to apoptosis in insects. In this embodiment, formulations of the invention would be applied to useful insects, e.g., bees and other insects that are involved in pollination of plants. In a specific embodiment, a formulation of the invention would be applied to bees involved in the production of honey. Generally, the methods described herein may be applied to any organism, e.g., eukaryote, that may have commercial importance. For example, they can be applied to fish (aquaculture) and birds (e.g., chicken and fowl).
Higher doses of-formulations of the invention may also be used as a pesticide by interfering with the regulation of silenced genes and the regulation of apoptosis during development. In this embodiment, a compound may be applied to plants using a method known in the art that ensures the compound is bio-available to insect larvae, and not to plants. At least in view of the link between reproduction and longevity (Longo and Finch, Science, 2002), formulations of the invention can be applied to affect the reproduction of organisms such as insects, animals and microorganisms.
4. Pharmaceutical Compositions
The pharmaceutical compositions described herein may be formulated in a conventional manner using one or more physiologically acceptable carriers or excipients. For example, compositions may be formulated for administration by, for example, injection (e.g. SubQ, IM, IP), inhalation or insufflation (either through the mouth or the nose) or oral, buccal, sublingual, transdermal, nasal, parenteral or rectal administration. In one embodiment, a formulation of the invention may be administered locally, at the site where the target cells are present, i.e., in a specific tissue, organ, or fluid (e.g., blood, cerebrospinal fluid, etc.).
In certain embodiments, the pharmaceutical compositions of the invention are administered orally as a liquid. In other embodiments, the pharmaceutical compositions of the invention are filled into capsules, such as soft gelatin capsules.
Pharmaceutical compositions can be formulated for a variety of modes of administration, including systemic and topical or localized administration. Techniques and formulations generally may be found in Remington's ' Pharmaceutical Sciences, Meade Publishing Co., Easton, PA. For parenteral administration, injection is preferred, including intramuscular, intravenous, intraperitoneal, and subcutaneous, preferably in physiologically compatible buffers such as Hank's solution or Ringer's solution.
Liquid preparations for oral administration may take the form of. for example, solutions, syrups or suspensions. Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives or hydrogenated edible fats); emulsifying agents (e.g., lecithin or acacia); non-aqueous vehicles (e.g., ationd oil, oily esters, ethyl alcohol or fractionated vegetable oils); and preservatives (e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid). The preparations may also contain buffer salts, flavoring, coloring and sweetening agents as appropriate. Preparations for oral administration may be suitably formulated to give controlled release of the active compound.
For administration by inhalation (e.g., pulmonary delivery), formulations of the invention may be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Pharmaceutical compositions may be foπnulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
In addition to the formulations described previously, formulations of the invention may be a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
In certain embodiments, the compounds described herein can be formulated for delivery to the central nervous system (CNS) (reviewed in Begley, Pharmacology & Therapeutics 104: 29-45 (2004)). Conventional approaches for drug delivery to the CNS include: neurosurgical strategies (e.g., intracerebral injection or intracerebroventricular infusion); molecular manipulation of the agent (e.g., production of a chimeric fusion protein that comprises a transport peptide that has an affinity for an endothelial cell surface molecule in combination with an agent that is itself incapable of crossing the BBB) in an attempt to exploit one of the endogenous transport pathways of the BBB; pharmacological strategies designed to increase the lipid solubility of an agent (e.g., conjugation of water-soluble agents to lipid or cholesterol carriers); and the transitory disruption of the integrity of the BBB by hyperosmotic disruption (resulting from the infusion of a mannitol solution into the carotid artery or the use of a biologically active agent such as an angiotensin peptide).
In one embodiment, a pharmaceutical composition described herein is a topical formulation containing a topical carrier that is generally suited to topical drug administration and comprising any such material known in the art. The topical carrier may be selected so as to provide the composition in the desired form, e.g., as an ointment, lotion, cream, microemulsion, gel, oil, solution, or the like, and may be comprised of a material of either naturally occurring or synthetic origin. It is preferable that the selected carrier not adversely affect the active agent or other components of the topical formulation. Examples of suitable topical carriers for use herein include water, alcohols and other nontoxic organic solvents, glycerin, mineral oil, silicone, petroleum jelly, lanolin, fatty acids, vegetable oils, parabens, waxes, and the like. Formulations may be colorless, odorless ointments, lotions, creams, microemulsions and gels.
Pharmaceutical compositions of the invention may be ointments, which generally are semisolid preparations which are typically based on petrolatum or other petroleum derivatives. The specific ointment base to be used, as will be appreciated by those skilled in the art, is one that will provide for optimum drug delivery, and, preferably, will provide for other desired characteristics as well, e.g., emolliency or the like. As with other carriers or vehicles, an ointment base should be inert, stable, nonirritating and nonsensitizing. As explained in Remington's (supra) ointment bases may be grouped in four classes: oleaginous bases; emulsifiable bases; emulsion bases; and water-soluble bases. Oleaginous ointment bases include, for example, vegetable oils, fats obtained from animals, and semisolid hydrocarbons obtained from petroleum. Emulsifiable ointment bases, also known as absorbent ointment bases, contain little or no water and include, for example, hydroxystearin sulfate, anhydrous lanolin and hydrophilic petrolatum. Emulsion ointment bases are either water-in-oil (W/O) emulsions or oil-in-water (O/W) emulsions, and include, for example, cetyl alcohol, glyceryl monostearate, lanolin and stearic acid. Exemplary water-soluble ointment bases are prepared from polyethylene glycols (PEGs) of varying molecular weight; again, reference may be had to Remington's, supra, for further information.
Pharmaceutical compositions of the invention may be creams, which generally are viscous liquid or semisolid emulsions, either oil-in-water or water-in- oil. Cream bases are water-washable, and contain an oil phase, an emulsifier and an aqueous phase. The oil phase is generally comprised of petrolatum and a fatty alcohol such as cetyl or stearyl alcohol; the aqueous phase usually, although not necessarily, exceeds the oil phase in volume, and generally contains a humectant. The emulsifier in a cream formulation, as explained in Remington 's, supra, is generally a nonionic, anionic, cationic or amphoteric surfactant.
Pharmaceutical compositions of the invention may be microemulsions, which generally are thermodynamically stable, isotropically clear dispersions of two immiscible liquids, such as oil and water, stabilized by an interfacial film of surfactant molecules (Encyclopedia of Pharmaceutical Technology (New York: Marcel Dekker, 1992), volume 9). For the preparation of microemulsions, surfactant (emulsifier), co-surfactant (co-emulsifier), an oil phase and a water phase are necessary. Suitable surfactants include any surfactants that are useful in the preparation of emulsions, e.g., emulsifiers that are typically used in the preparation of creams. The co-surfactant (or "co-emulsifer") is generally selected from the group of polyglycerol derivatives, glycerol derivatives and fatty alcohols. Preferred emulsifier/co-emulsifier combinations are generally although not necessarily selected from the group consisting of: glyceryl monostearate and polyoxyethylene stearate; polyethylene glycol and ethylene glycol palmitostearate; and caprilic and capric triglycerides and oleoyl macrogolglycerides. The water phase includes not only water but also, typically, buffers, glucose, propylene glycol, polyethylene glycols, preferably lower molecular weight polyethylene glycols (e.g., PEG 300 and PEG 400), and/or glycerol, and the like, while the oil phase will generally comprise, for example, fatty acid esters, modified vegetable oils, silicone oils, mixtures of mono- di- and triglycerides, mono- and di-esters of PEG (e.g., oleoyl macrogol glycerides), etc.
Pharmaceutical compositions of the invention may be gel formulations, which generally are semisolid systems consisting of large organic molecules distributed substantially uniformly throughout a carrier liquid (single phase gels). Single phase gels can be made, for example, by combining the active agent, a carrier liquid and a suitable gelling agent such as tragacanth (at 2 to 5%), sodium alginate (at 2-10%), gelatin (at 2-15%), methylcellulose (at 3-5%), sodium carboxymethylcellulose (at 2-5%), carbomer (at 0.3-5%) or polyvinyl alcohol (at 10-20%) together and mixing until a characteristic semisolid product is produced. Other suitable gelling agents include methylhydroxycellulose, polyoxyethylene- polyoxypropylene, hydroxyethylcellulose and gelatin. Although gels commonly employ aqueous carrier liquid, alcohols and oils can be used as the carrier liquid as well.
Various additives, known to those skilled in the art, may be included in formulations, e.g., topical formulations. Examples of additives include, but are not limited to, solubilizers, skin permeation enhancers, opacifiers, preservatives (e.g., anti-oxidants), gelling agents, buffering agents, surfactants (particularly nonionic and amphoteric surfactants), emulsifiers, emollients, thickening agents, stabilizers, humectants, colorants, fragrance, and the like. Inclusion of solubilizers and/or skin permeation enhancers is particularly preferred, along with emulsifiers, emollients and preservatives. An optimum topical formulation comprises approximately: 2 wt. % to 60 wt. %, preferably 2 wt. % to 50 wt. %, solubilizer and/or skin permeation enhancer; 2 wt. % to 50 wt. %, preferably 2 wt. % to 20 wt. %, emulsifiers; 2 wt. %- to 20 wt. % emollient; and 0.01 to 0.2 wt. % preservative, with the active agent and earner (e.g.. water) making of the remainder of the formulation.
A skin permeation enhancer serves to facilitate passage of therapeutic levels of active agent to pass through a reasonably sized area of unbroken skin. Suitable enhancers are well known in the art and include, for example: lower alkanols such as methanol ethanol and 2-propanol; alkyl methyl sulfoxides such as dimethylsulfoxide (DMSO), decylmethylsulfoxide (Cm MSO) and tetradecylmethyl sulfboxide; pyrrolidones such as 2-pyrrolidone, N-methyl-2-pyrrolidone and N-(- hydroxyethyl)pyπolidone; urea; N,N-diethyl-m-toluamide; C? -C6 alkanediols; miscellaneous solvents such as dimethyl formamide (DMF), N.N- dimethylacetamide (DMA) and tetrahydrofurfuryl alcohol; and the 1 -substituted azacycloheptan-2-ones, particularly 1 -n-dodecylcyclazacycloheptan-2-one (laurocapram; available under the trademark AzoneR™ from Whitby Research Incorporated, Richmond, Va.).
Examples of solubilizers include, but are not limited to, the following: hydrophilic ethers such as diethylene glycol monoethyl ether (ethoxydiglycol, available commercially as TranscutolRTM) and diethylene glycol monoethyl ether oleate (available commercially as SoftcutolRTM); polyethylene castor oil derivatives such as polyoxy 35 castor oil, polyoxy 40 hydrogenated castor oil, etc.; polyethylene glycol, particularly lower molecular weight polyethylene glycols such as PEG 300 and PEG 400, and polyethylene glycol derivatives such as PEG-8 caprylic/capric glycerides (available commercially as LabrasolRTM); alkyl methyl sulfoxides such as DMSO; pyrrolidones such as 2-pyrrolidone and N-methyl-2- pyrrolidone; and DMA. Many solubilizers can also act as absorption enhancers. A single solubilizer may be incorporated into the formulation, or a mixture of solubilizers may be incorporated therein. In certain embodiments, pharmaceutical compositions of the invention exclude solubilizers that act as permeation enhancers, particularly ethoxydiglycol.
Suitable emulsifiers and co-emulsifiers include, without limitation, those emulsifiers and co-emulsifiers described with respect to microemulsion formulations. Emollients include, for example, propylene glycol, glycerol, isopropyl myristate, polypropylene glycol-2 (PPG-2) myristyl ether propionate, and the like.
Other active agents may also be included in formulations, e.g., other antiinflammatory agents, analgesics, antimicrobial agents, antifungal agents, antibiotics, vitamins, antioxidants, and sunblock agents commonly found in sunscreen formulations including, but not limited to, anthranilates, benzophenones (particularly benzophenone-3), camphor derivatives, cinnamates (e.g., octyl methoxycinnamate), dibenzoyl methanes (e.g., butyl methoxydibenzoyl methane), p-aminobenzoic acid (PABA) and derivatives thereof, and salicylates (e.g., octyl salicylate). In certain topical formulations, the active agent is present in an amount in the range of approximately 0.25 wt. % to 75 wt. % of the formulation, preferably in the range of approximately 0.25 wt. % to 30 wt. % of the formulation, more preferably in the range of approximately 0.5 wt. % to 15 wt. % of the formulation, and most preferably in the range of approximately 1.0 wt. % to 10 wt. % of the formulation.
Topical skin treatment compositions can be packaged in a suitable container to suit its viscosity and intended use by the consumer. For example, a lotion or cream can be packaged in a bottle or a roll-ball applicator, or a propellant-driven aerosol device or a container fitted with a pump suitable for finger operation. When the composition is a cream, it can simply be stored in a non-deformable bottle or squeeze container, such as a tube or a lidded jar. The composition may also be included in capsules such as those described in U.S. Pat. No. 5,063,507.
Accordingly, also provided are closed containers containing a cosmetically acceptable composition as herein defined.
In an alternative embodiment, a pharmaceutical formulation is provided for oral or parenteral administration, in which case the formulation may comprises a modulating compound-containing microemulsion as described above, but may contain alternative pharmaceutically acceptable carriers, vehicles, additives, etc. particularly suited to oral or parenteral drug administration. Alternatively, a microemulsion may be administered orally or parenterally substantially as described above, without modification. Pharmaceutical compositions described herein may be stored in oxygen free environment according to methods in the art. For example, resveratrol or analog thereof can be prepared in an airtight capsule for oral administration, such as Capsugel from Pfizer, Inc.
The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such compounds may lie within a range of circulating concentrations that include the EDso with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the ICso (i.e., the concentration of the test compound that achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography.
5. Kits
Also provided herein are kits, e.g., kits for therapeutic purposes or kits for modulating the lifespan of cells or modulating apoptosis. A kit may comprise one or more pharmaceutical compositions of the invention, e.g., in premeasured doses. A kit may optionally comprise devices for contacting cells with the compounds and instructions for use. Devices include syringes, stents and other devices for introducing pharmaceutical compositions of the invention into a subject (e.g., the blood vessel of a subject) or applying it to the skin of a subject.
In yet another embodiment, the invention provides a composition of matter comprising a formulation of this invention and another therapeutic agent (the same ones used in combination therapies and combination compositions) in separate dosage forms, but associated with one another. The term "associated with one another" as used herein means that the separate dosage forms are packaged together or otherwise attached to one another such that it is readily apparent that the separate dosage forms are intended to be sold and administered as part of the same regimen. The agent and the formulation of the invention are preferably packaged together in a blister pack or other multi-chamber package, or as connected, separately sealed containers (such as foil pouches or the like) that can be separated by the user (e.g., by tearing on score lines between the two containers).
In still another embodiment, the invention provides a kit comprising in separate vessels, a) a formulation of the invention; and b) another another therapeutic agent such as those described elsewhere in the specification.
The practice of the present methods will employ, unless otherwise indicated, conventional techniques of cell biology, cell culture, molecular biology, transgenic biology, microbiology, recombinant DNA, and immunology, which are within the skill of the art. Such techniques are explained fully in the literature. See, for example, Molecular Cloning A Laboratory Manual, 2nd Ed., ed. by Sambrook, Fritsch and Maniatis (Cold Spring Harbor Laboratory Press: 1989); DNA Cloning, Volumes I and II (D. N. Glover ed., 1985); Oligonucleotide Synthesis (M. J. Gait ed., 1984); Mullis et al. U.S. Patent No: 4,683,195; Nucleic Acid Hybridization (B. D. Hames & S. J. Higgins eds. 1984); Transcription And Translation (B. D. Hames & S. J. Higgins eds. 1984); Culture Of Animal Cells (R. I. Freshney, Alan R. Liss, Inc., 1987); Immobilized Cells And Enzymes (IRL Press, 1986); B. Perbal, A
Practical Guide To Molecular Cloning (1984); the treatise, Methods In Enzymology (Academic Press, Inc., N. Y.); Gene Transfer Vectors For Mammalian Cells (J. H. Miller and M. P. Calos eds., 1987, Cold Spring Harbor Laboratory); Methods In Enzymology, VoIs. 154 and 155 (Wu et al. eds.), Immunochemical Methods In Cell And Molecular Biology (Mayer and Walker, eds., Academic Press, London, 1987); Handbook Of Experimental Immunology, Volumes I-1V (D. M. Weir and C. C. Blackwell, eds., 1986); Manipulating the Mouse Embryo, (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1986).
EXEMPLIFICATION
The invention now being generally described, it will be more readily understood by reference to the following examples which are included merely for purposes of illustration of certain aspects and embodiments of the present invention, and are not intended to limit the invention in any way.
Resveratrol formulation development
Lipid Matrix formulation:
A liquid formulation was prepared by dissolving resveratrol (drug substance,
DS) and monoacyl phospholipid in a mixture of PEG 300 and propyleneglycol. The ratio between the DS and phospholipid was 1/12 w/w in PEG 300/ propyleneglycol
1/1 w/w as solvent mixture. The formulation, which has a 5% drug load, was clear after dilution in purified water for more than 4 hours.
Emulsion Drug Delivery formulations: Two formulations were prepared. One formulation comprises a surfactant mixture forming mixed micelles after dilution with purified water. The first formulation (Formulation 13) has a 5% drug load, contains 45 % polyethyoxylated glycol hydroxystearate (Solutol™HS15), 5 % PEG-ylated Vitamin E (Vit E TPGS) in a solvent mixture of PEG 300 and EtOH 1/1 w/w.
The second formulation (Formulation 14) forms a micro-emulsion after dilution with purified water and has a 5% drug load, contains 23.75 % medium chain triglyceride (Mygliol 812N), 42.75 % polyethoxylated hydrogenated castor oil (Cremophor™ RH40), 4.75% Glyceryl MonoOleate (Peceol™) and 23.75 % anhydrous ethanol as solvent.
Polymer formulation: The Drug Substance (DS) has a high affinity for polymers relative to other excipients. Polyvinylpyrrolidone (PVP) was found to be a suitable polymer to delay the drug precipitation upon dilution of formulations in which the DS was dissolved, with water. Solid dispersions were produced by solubilizing the DS polymer and surfactant (monoacyl phospholipid or Vit E TGPS or Labrasol™) in a solvent followed by removal of the solvent. In Table 3, below, the compositions of the explored solid dispersion type of formulation with resveratrol and the DS precipitation tendency of the formulation upon dilution with water are provided.
In Table 3, it is shown that resveratrol could be solubilized when a sufficient amount of polymer was used in presence of a minimum amount of surfactant. Formulation 2 and Formulation 3 compare the effect of the DS to lipid ratio at the same DS to polymer ratio; Formulation 3, Formulation 4 and Formulation 5 compare the effect of decreasing the polymer to DS ratio at a constant DS to lipid ratio.
Table 3: Composition of solid dispersions with resveratrol (Drug Substance (DS) and tendency of precipitation upon hydration/dilution of the formulations in purified water at
RT.
* Precipitates are not crystalline
In order to obtain a formulation with fully solubilized drug upon hydration of the solid dispersion, a maximum drug load of 7 % was achievable (Formulation 3 for having a fully solubilized DS). When DS precipitates upon hydration of formulations containing PVP polymer, the drug did not precipitate in a crystalline/needle form as was observed under the microscope (x 400) but in a noncrystalline, probably amorphous form. This sort of amorphous precipitates, obtained after hydration of such formulation could demonstrate higher oral bioavailability. This means that solid dispersions containing considerably more that the studied range of up to 10 % DS could give rise to a sufficient degree of oral absorption.
High Concentration Formulations
Starting from Formulation 13, the drug load was increased to 10% using the same 1/1 w/w ratio of Vit E TPGS/DS and replacing propyleneglycol with Labrasol™. Upon one-fold dilution with water a slight DS precipitation was observed.
Formulation 14 comprised Medium Chain Triglycerides and a 10% drug load (see full composition below), but showed a fast precipitation of the DS upon one- fold dilution with purified water. Formulation 1 1 comprised Long Chain Triglyceride (Soya oil), Capryol™ ■ 90, Tween™ 80, Gelucire™ 44/14 and ethanol, and showed hardly any precipitation upon one-fold dilution with purified water. After a few hours, some oil droplets were visible on the surface of the water phase. To further improve formulation Formulation 1 1 , the use of the two lipids
Gelucire™ 44/14 and Labrasol™ were explored as main components in the formulations. It was found that when DS was dissolved in Gelucire™ 44/14 at 100 mg/g, the drug precipitated in a fast manner after dilution with purified water. At a concentration of 150 mg DS/g In Labrasol™ (Formulation 10), no precipitation of crystalline drug was observed upon dilution with purified water. The DS precipitated in the form of oil-like droplets. With a combination of both excipients, a drug load of 10% DS could be achieved without the need of ethanol. Higher drug loads can be achieved by using the two lipids in combination with ethanol as additional solvent/excipient to fully solubilize the drug in the formulation. Based on the results of Table 4, the Formulation 10 showed an increase of degradation product of 0.1 % and 0.5% after 1 -week storage at 25°C and 400C, respectively. It is important to note that the color of the formulation changed from a slight yellowish color at TO to a slight pink and dark red at 25°C and 400C, respectively. Formulation 1 1 also showed a color change at 25°C and 400C compared to the initial color but no increase of degradation products was observed.
Table 4: Stability study results of formulations #10 and #1
Degradation Products 0.2% 0.2%
Observation Yellow Brown turbid turbid liquid liquid
! ) Value at TO was measured on frozen samples at -18°C; 2) Initial drug substance contains already 0.2% of degradation products
A summary of some of the resveratrol formulations is shown in Table 5 below.
Table 5. Resveratrol Solution-based Formulations
In Vivo Studies with resveratrol formulations Mouse studies
The objective of this study was to assess the pharmacokinetics of resveratrol in 3 formulations in C57BL/6 mice following oral administration. For this purpose, 72 C57BL/6 mice were divided into 3 treatment groups, 24 per group. Test compound resveratrol was dissolved in vehicles and orally administrated to those mice at 500 mg/kg. Serial plasma samples were collected following oral administration, after which resveratrol concentration in plasma were determined and pharmacokinetic property in mice were evaluated.
Preparation of Formulation 12
400 mg of resveratrol were weighed into a mortar and pestle for grinding. To this was added 7.6 g of lipid matrix vehicle 1 (60% monoacyl phospholipid, 20% PEG300, 20% propyleneglycol) and the contents were stirred rapidly. After stirring homogenously, 0.9% saline was added to lower viscosity and the container was rinsed with 0.9% saline, and the mixture was vortexed thoroughly to yield a homogenous suspension at 25 mg/mL. Animals were dosed orally at 20 ml/kg to achieve an oral dose of 500 mg/kg.
Preparation of Formulation 13
400 mg of resveratrol were weighed into a vial for mixing. 7.6 g from the vial labeled emulsion vehicle (45% polyethyoxylated glycol hydroxystearate (Solutol™ HS 15); 5.0% PEG-ylated Vitamin E (Vit E TPGS); 50% PEG300/ EtOH (1/1 w/w) were added and the mixture was vortexed until complete dissolution of the drug substance at 50 mg/mL by ultrasonic. Animals were dosed orally at 10 ml/kg to achieve an oral dose of 500 mg/kg.
Preparation of Formulation 14
400 mg resveratrol were weighed into a vial for mixing. 7.6 g from the vial labeled lipid matrix vehicle 2 (25% medium chain triglyceride (Mygliol ™ 812N); 45% polyethoxylated hydrogenated castor oil (Cremaphor™ RH40); 5.0% Glycerl MonoOleate (Peceol™); 25% ethanol) were added and the mixture was vortexed until complete dissolution of the drug substance at 50 mg/mL by ultrasonic. Animals were dosed orally at 10 ml/kg to achieve an oral dose of 500 mg/kg.
13 Blood samples were collected by eyeball extirpation at 5, 15, 30, 60, 240, and 360 min post-dose. These samples were then centrifuged at 8,000 rpm (RCF was 4262 X g) for 6 min and the resultant plasma transferred into plain Eppendorff tubes and deeply frozen at -20°C until bioanalysis.
Analysis
The concentrations of resveratrol in plasma were determined using a high performance liquid chromatography/mass spectrometry (HPLC/MS/MS) method. Plasma samples were processed by liquid-liquid extraction. Plasma samples (0.1 niL) were transferred to Eppendorff tube, then 20 μL methanol, 50 μL internal standard solution (1 μg/mL) and 500 μL methyl tert-butyl ether (MTBE) were added to it. Internal standard solution was prepared by dissolving IS compound (CAS: 2896-608) in H2O to yield a final concentration of 200 μg/mL, then diluted to 1 μg/mL. After vortexing for 1 min and centrifuging for 5 min at 15,000 rpm, 450 μL of supernatant was transferred to a glass tube and evaporated under vacuum condition. The residue was reconstituted with 100 μL methanol
LC-MS/MS Apparatus The LC system comprised an Agilent (Agilent Technologies Inc. USA) liquid chromatograph equipped with an isocratic pump ( 1 100 series), an autosampler ( 1 100 series) and a degasser (1 100 series). Mass spectrometric analysis was performed using an AP13000 (triple-quadrupole) instrument from AB Inc (Canada) with an ESI interface. The data acquisition and control system were created using Analyst 1.4 software from ABI Inc.
Chromatographic Conditions
Column: Waters Symmetry 5 μm Cl 8 ( 150 mmχ2. 1 mm) Mobile phase: H2O : Methanol = 10: 90 Elution rate: 250 μL/Min Column temperature: 25°C Injection volume: 5 μL Mass
Scan type: Negative MRM Ion source Turbo spray Ionization model ESI Nebulize gas: 8 L/min Curtain gas: 8 L/min Collision gas: 4 L/min Ionspray voltage: -4500 V; Temperature: 450°C
Following oral administration of 3 vehicles of resveratrol at a dose of 500mg/kg, the pharmacokinetic profile was as below and in Table 6.
For Formulation 12, the values of C1113x and T1113x for resveratrol were 7046.01 μg/L, 0.25 h, respectively, while the values of AUQo-t) and Ti/? were 4774.71 h*μg/L and 1.27 h, respectively.
For Formulation 13, the values of C1113x and T1113x for resveratrol were 9145.49 μg/L, 0.08 h, respectively, while the values of AUQo-t) and Ti/2 were 5457.66 h*μιg/L and 4.08 h, respectively. For Formulation 14, the values of Cmax and T1113x for resveratrol were 5431 .25 μg/L, 0.08 h, respectively, while the values of AUQo-o and T\a were 8325.98 h*μg/L and 1 .60 h, respectively.
Table 6. Selected Pharmacokinetics Parameters of resveratrol in Male C57BL/6 Mice Following Oral Administration in 3 Vehicles
AUC(o-t) AUC(o-∞) t|/2 T 1 max *— max ug/L*h ug/L*h h h ug/L
Formulation
4774.71 4996.40 1.27 0.25 7046.01
12
Formulation
5457.66 7473.03 4.08 0.08 9145.49 13
Formulation
8325.98 9088.90 1 .60 0.08 5431 .25 14
Rat studies The objective of this study was to assess the pharmacokinetics of resveratrol in formulations in rats following oral administration. For this purpose, 72 Sprague
Dawley rats (males, 8- 10 weeks old, 200 to 250gr) were divided into 4 treatment groups, 18 per group. Test compound resveratrol was dissolved in vehicles and orally administrated to the rats at 100 or 500 mg/kg. Serial plasma samples were collected following oral administration, after which resveratrol concentration in plasma were determined and pharmacokinetic property in mice were evaluated.
Formulation 10 and 15
Dose Preparation and Analysis and Dose Administration.
Compounds were dissolved in either Labrasol (Formulation 10) or Labrasol/ Lecithin (33/1 v/v; Formulation 15) to yield a final concentration at 10 mg/mL and 50 mg/mL (free base) for a dosing volume of 10 mL/kg. Dose volumes for each test animal were determined based on individual body weight taken on the day of dosing. Dosing concentration was reconfirmed by HPLC.
Blood Sample Collection
The blood was collected at 0 (pre-dosing) and 5 min, 15 min, 30 min, 1 hr, 2 hr, 4 hr, 6 hr, 8 hr and 24 hr post administration. Blood samples for each time point were collected within 10% for the time points before the first hour and within ±2 min for the time points after 1 hour. The plasma was separated via centrifugation within 1 hr and stored in -20°C before analysis. Analysis was done as before for the mouse study in the previous example. Results are shown in Table 7.
Table 7. Selected Pharmacokinetics Parameters of Resveratrol in Rats Following Oral Administration in Two Formulations at 100 and 500 mg/kg
Pharmacokinetic Parameters
100 n-m/ka
AUQo-O AUC(o-.β) MRT(O- ) t|/2z T 1 max ^ιπ;i.\ μg/L*hr μg/L*hr hr hr hr μg/L
Formulation 10
1 2379.71 2800.27 3.53 2. 14 0.08 1862.37
2 1 555.61 1 81 1 .28 3.52 2.1 8 0.08 1392.59
3 1720.56 1926. 1 2 3.37 2.1 7 0.08 1836.78 mean 1 885.30 21 79.22 3.47 2. 16 0.08 1697.25
SD 436.05 540.90 0.09 0.02 0.00 264.15
Formulation 15
4 1684.63 1 850.09 2.84 1 .07 0.25 1 180.57 5 1388.70 1520.22 2.69 1.94 0.08 1303.38
6 1216 .34 1340.39 3 .14 2.18 0.08 714.44 mean 1429 .89 1570.24 2 .89 1.73 0.14 1066.13
SD 236. 85 258.50 0 .23 0.58 0.10 310.70
500 mg/kg
AUC(O-.) AUC(o-oo) MRT(o-) tl/2z T 1 max Cinax μg/L*hr μg/L*hr hr hr hr μg/L
Formulation 10
1 28824.97 30192.73 7.01 5.55 0.08 4152.03
2 15075.47 18551.24 13.28 10.23 0.08 4417.54
3 15095.18 20978.36 18.19 14.23 0.08 4010.32 mean 19665.20 23240.77 12.83 10.00 0.08 4193.30
SD 7932.60 6141.66 5.61 4.34 0.00 206.72
Formulation 15
4 12881.60 13760.14 8.79 6.20 0.08 2103.65
5 19861.17 2181 1.59 9.79 6.88 0.08 3691.06
6 17873.80 19430.63 9.03 6.62 0.08 3682.48 mean 16872.19 18334.12 9.20 6.57 0.08 3 159.06
SD 3595.97 4136.21 0.52 0.35 0.00 914.02
Human Studies
Formula 15 was prepared as follows. Resveratrol (99% Grade from PCAS, Lima France) was filled into 00 capsules (Torpac USP Grade gelatin capsules) using a Profill Capsule Filling System (Torpac, Model No. FB-2783). 100 capsules were flush filled with resveratrol according to the filling system operating instructions. The resulting filled capsules were visually inspected and weighed. Average net capsule weight was 544.1 mg/capsule (standard deviation was 16.6 mg/capsule). 5 capsules are approximately the equivalent of 2.7 grams of resveratrol.
Preparation of Formula 10 was done as described above. Resveratrol (99% Grade from PCAS, Lima France) was dissolved in Caprylocapryol Macrogol glycerides (Labrasol™) at a final concentration of 15% w/w. A 2.7 gram dose was approximately 18 mis.
Formula 10 was also prepared as soft gel capsules (Formula 10a). The liquid capsules for dosing were produced by manufacturing a 300 g batch of the resveratrol fill formulation, filling that formulation into liquid capsules and then sealing the capsules. Specifically, 30% resveratrol was added to 70% caprylocaproyl macrogol 8 glycerides (Labrasol, Gattefosse) and mixed at 400C for 60 minutes (all percentages are on a weight basis). The resulting fill formulation is an opaque, homogenous suspension. The fill formulation was continually mixed while cooling to 300C and then transferred to the filling hopper for capsule filling. Specific gravity of the mixture was 1.1 (at 25°C).
For the capsule filling and sealing, the fill product hopper temperature was set at 30°C, and the drying air temperature was set to 400C. Liquid gelatin capsules (Size 00 Licaps HGC Natural/Natural, Capsugel) were filled and sealed on a CFS 1200 Capsule Filling and Sealing Machine (Capsugel) according to the equipments standard operating procedures. The capsules were filled volumetrically with 0.75 ± 0.10 mL of the fill formulation. The theoretical concentration of resveratrol in each capsule was 250 mg/capsule. Seven capsules were orally dosed to delivery 1.7 g of active resveratrol. The study was carried out in 4 subjects who received different resveratrol formulations by oral administration. Administration of resveratrol ( 1.7 or 2.7 g) took place approximately 15 min after subjects ingested a standard breakfast. Blood samples were collected to assess the PK parameters of each formulation at the following time points: Baseline (pre-administration), 30 and 60 minutes, 2 hr, 4 hr and 24 hr. Blood samples for PK analysis were collected in EDTA vacutainers (~5 mL per draw).
Plasma concentrations of resveratrol were estimated using a validated LCMS/MS method. Tmax, Cmax, and AUC(0-4hr) were evaluated using standard statistical software. Results are shown in Table 8 and Figure 1. Formulation 10 provides significant exposure (287% increase in AUC and 186% in Cmax) of Resveratrol compared to the reference Resveratrol capsule. Table 8. Selected Pharmacokinetics Parameters of Resveratrol in Humans Following Oral Administration in Three Formulations at Indicated Dose
EQUIVALENTS The present invention provides among other things sirtuin-activating compounds and methods of use thereof. While specific embodiments of the subject invention have been discussed, the above specification is illustrative and not restrictive. Many variations of the invention will become apparent to those skilled in the art upon review of this specification. The full scope of the invention should be deteπnined by reference to the claims, along with their full scope of equivalents, and the specification, along with such variations.
INCORPORATION BY REFERENCE
All publications and patents mentioned herein, including those items listed below, are hereby incorporated by reference in their entirety as if each individual publication or patent was specifically and individually indicated to be incorporated by reference. In case of conflict, the present application, including any definitions herein, will control.
Also incorporated by reference in their entirety are any polynucleotide and polypeptide sequences which reference an accession number correlating to an entry in a public database, such as those maintained by The Institute for Genomic Research (TIGR) (www.tigr.org) and/or the National Center for Biotechnology Information (NCBl) (www.ncbi.nlm.nih.gov).
Also incorporated by reference are the following: PCT Publications WO 2005/002672; 2005/002555; and 2004/016726.

Claims

CLAIMSWhat is claimed is:
1. An oral pharmaceutical composition comprising a solution or emulsion comprising at least 10% by weight of resveratrol or ester thereof.
2. The composition of claim 1 , wherein the solution or emulsion comprises at least 15% by weight of resveratrol.
3. The composition of claim 1 , wherein said solution or emulsion comprises resveratrol and one or more of monoglycerides, diglycerides, triglycerides, fatty acid monoesters of polyethylene glycol (PEG) and fatty acid diesters of PEG.
4. The composition of claim 3, wherein the monoglycerides, diglycerides, triglycerides, fatty acid monoesters of PEG and fatty acid diesters of PEG include C8 and C l O fatty acids.
5. The composition of claim 4, wherein the solution comprises resveratrol and caprylocapryol macrogol glycerides.
6. The composition of claim 3, further comprising a polar lipid.
7. The composition of claim 6, wherein the polar lipid is a phospholipid.
8 The composition of claim 7, wherein the phospholipid is lecithin.
9. The composition of claim 3, further comprising one or more of saturated polyglycolized glycerides and ethanol.
10. The composition of claim 3, wherein the composition is a solution.
1 1. The composition of claim 10, wherein the solution consists essentially of resveratrol and caprylocapryol macrogol glycerides.
12. The composition of claim 1 , wherein the solution or emulsion comprises a triglyceride, a propylene glycol fatty acid ester, a polyethylene glycol sorbitan fatty acid ester, a saturated polyglycolized glyceride and an alcohol.
13. The composition of claim 12, wherein the triglyceride is a long chain triglyceride, the propylene glycol fatty acid ester is propylene glycol caprylate/caprate, the polyethylene glycol sorbitan fatty acid ester is polyoxyethylated sorbitan monooleate, the saturated polyglycolized glyceride is lauryol macrogol glycerides and the alcohol is ethanol.
14. The composition of claim 1 , wherein the solution or emulsion comprises resveratrol, a monoacyl phospholipid, a PEG and a dihydric alcohol.
15. The composition of claim 14, wherein the PEG is PEG 300 and the dihydric alcohol is propylene glycol.
16. The composition of claim 1 , wherein the solution or emulsion comprises resveratrol, a polyethoxylated fatty acid, an alcohol-oil transesterification product and an alcohol.
17. The composition of claim 16, wherein the polyethoxylated fatty acid is polyethoxylated glycol hydroxystearate, the alcohol-oil transesterification product is D-alpha-tocopheryl PEG 1000 succinate and the alcohol is ethanol.
18. The composition of claim 1 , wherein the solution or emulsion comprises resveratrol, a triglyceride, an alcohol-oil transesterification product, a monoglyceride and an alcohol.
19 The composition of claim 18, wherein the triglyceride is a medium chain triglyceride, the alcohol-oil transesterification product is a polyethxoylated hydrogenated castor oil, the monoglyceride is an oleate and the alcohol is ethanol.
20. The composition of claim 1 , comprising a polymer and a surfactant.
21. The composition of claim 20, wherein the polymer is an amine-containing polymer.
22. The composition of claim 21 , wherein the amine-containing polymer is polyvinylpyrrolidone.
23. A method of preparing the pharmaceutical composition of claim 1 , comprising mixing resveratrol and one or more of monoglycerides, diglycerides, triglycerides, fatty acid monoesters of polyethylene glycol (PEG) and fatty acid diesters of PEG.
24. The method of claim 23, wherein the method comprises mixing resveratrol and caprylocapryol macrogol glycerides.
25. A method of preparing the pharmaceutical composition of claim 1 , comprising mixing resveratrol and a triglyceride, a propylene glycol fatty acid ester, a polyethylene glycol sorbitan fatty acid ester, a saturated polyglycolized glyceride and an alcohol.
26. A method of preparing the pharmaceutical composition of claim 1 , comprising mixing resveratrol and a monoacyl phospholipid, a PEG and a dihydric alcohol.
27. A method of preparing the pharmaceutical composition of claim 1, comprising mixing resveratrol and a polyethoxylated fatty acid, an alcohol- oil transesterification product and an alcohol.
28. A method of preparing the pharmaceutical composition of claim 1 , comprising mixing resveratrol and a triglyceride, an alcohol-oil transesterification product, a monoglyceride and an alcohol.
29. A method of preparing the pharmaceutical composition of claim 1 , comprising mixing resveratrol and a polymer and a surfactant.
EP09701250A 2008-01-08 2009-01-07 Resveratrol formulations Withdrawn EP2249806A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US1050508P 2008-01-08 2008-01-08
PCT/US2009/000078 WO2009089011A2 (en) 2008-01-08 2009-01-07 Resveratrol formulations

Publications (1)

Publication Number Publication Date
EP2249806A2 true EP2249806A2 (en) 2010-11-17

Family

ID=40522336

Family Applications (1)

Application Number Title Priority Date Filing Date
EP09701250A Withdrawn EP2249806A2 (en) 2008-01-08 2009-01-07 Resveratrol formulations

Country Status (3)

Country Link
US (1) US20110009496A1 (en)
EP (1) EP2249806A2 (en)
WO (1) WO2009089011A2 (en)

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102083438B (en) 2008-05-01 2016-11-09 西特里斯药业公司 The quinoline of conditioning agent and related analogs as silent message regulatory factor
EA020578B1 (en) 2008-07-03 2014-12-30 Сертрис Фармасьютикалз, Инк. Benzimidazoles and related analogs as sirtuin modulators
AU2009295948B2 (en) 2008-09-29 2013-12-05 GlaxoSmithKline, LLC Quinazolinone, quinolone and related analogs as sirtuin modulators
CA2755069A1 (en) 2009-03-16 2010-09-23 Genmedica Therapeutics Sl Anti-inflammatory and antioxidant conjugates useful for treating metabolic disorders
SG176571A1 (en) * 2009-05-19 2012-01-30 Vivia Biotech Sl Methods for providing personalized medicine tests ex vivo for hematological neoplasms
CN101579291B (en) * 2009-05-20 2011-06-15 清华大学 Resveratrol phospholipid composite nano-emulsion and preparation method and application thereof
BR112012010010A2 (en) 2009-10-29 2018-03-20 Sirtris Pharmaceuticals Inc bicyclic pyridines and analogues as modulators of sirtuin
US20120058088A1 (en) * 2010-06-28 2012-03-08 Resveratrol Partners, Llc Resveratrol-Containing Compositions And Methods Of Use
WO2012114204A2 (en) * 2011-02-15 2012-08-30 Ecole Polytechnique Federale De Lausanne (Epfl) Epfl-Tto Methods of treating mitochondrial dysfunction
CN102188370B (en) * 2011-04-27 2013-07-24 中国人民解放军第二军医大学 Resveratrol injection solution and intravenous injection
US8916528B2 (en) 2011-11-16 2014-12-23 Resveratrol Partners, Llc Compositions containing resveratrol and nucleotides
EP2788088B1 (en) 2011-12-06 2019-06-19 Unilever Plc. Skin anti-ageing composition
GB201217296D0 (en) * 2012-09-27 2012-11-14 Alta Innovations Ltd Method of treatment and/or prevention
DE102012025005A1 (en) * 2012-12-20 2014-06-26 Herbalist & Doc Gesundheitsgesellschaft Mbh Use of resveratrol and/or their analogues e.g. for treating platelet aggregation and cancellation of resistance to acetylsalicylic acid or nonsteroidal anti-inflammatory drugs and/or repealing paradoxical reaction to acetylsalicylic acid
US9724281B2 (en) * 2015-01-20 2017-08-08 Bruce L. Howe Composition of resveratrol and method for its use in the treatment of skin conditions
US20190046466A1 (en) * 2015-08-20 2019-02-14 Paul A. Knepper Composition and method for inhibiting platelet aggregation
US11318110B2 (en) 2015-08-28 2022-05-03 Caliway Biopharmaceuticals Co., Ltd. Pharmaceutical composition for reducing local fat and uses thereof
KR102240221B1 (en) * 2015-08-28 2021-04-14 칼리웨이 바이오파마슈티칼스 코., 엘티디. Pharmaceutical composition used for reducing localised fat and use of pharmaceutical composition
WO2021102361A1 (en) * 2019-11-22 2021-05-27 The Regents Of The University Of California Caspase 6 inhibitors and uses thereof
WO2021102299A1 (en) * 2019-11-22 2021-05-27 The Regents Of The University Of California Caspase 6 inhibitors and uses thereof
WO2022132844A1 (en) * 2020-12-15 2022-06-23 Muhammed Majeed Anti-viral compositions
EP4019013A1 (en) * 2020-12-24 2022-06-29 Bionotus GCV Treatment of pulmonary disorders
CN114223849A (en) * 2021-12-28 2022-03-25 青岛胡子叔叔食品科技有限公司 Instant chicken breast meat assisting in reducing fat and generating ketone and preparation method thereof
EP4212151A3 (en) * 2022-01-13 2023-09-27 Paul A. Knepper Stilbene, flavonol or curcumin compounds for use in the treatment of age-related macular degeneration (amd; armd)

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6414037B1 (en) * 1998-01-09 2002-07-02 Pharmascience Pharmaceutical formulations of resveratrol and methods of use thereof
US6008260A (en) * 1998-01-09 1999-12-28 Pharmascience Cancer chemopreventative composition and method
US7030155B2 (en) * 1998-06-05 2006-04-18 Sonus Pharmaceuticals, Inc. Emulsion vehicle for poorly soluble drugs
AU2006249816A1 (en) * 2005-05-25 2006-11-30 Sirtris Pharmaceuticals, Inc. Treatment of eye disorders with sirtuin modulators

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2009089011A2 *

Also Published As

Publication number Publication date
WO2009089011A8 (en) 2010-08-26
WO2009089011A2 (en) 2009-07-16
WO2009089011A3 (en) 2009-09-03
US20110009496A1 (en) 2011-01-13

Similar Documents

Publication Publication Date Title
US20110009496A1 (en) Resveratrol formulations
US7998974B2 (en) Fused heterocyclic compounds and their use as sirtuin modulators
EP1910362B9 (en) Imidazopyridine derivatives as sirtuin modulating agents
AU2006269459B2 (en) Methods and related compositions for treating or preventing obesity, insulin resistance disorders, and mitochondrial-associated disorders
US20090069301A1 (en) Acridine and Quinoline Derivatives as Sirtuin Modulators
US20090163476A1 (en) N-Phenyl Benzamide Derivatives as Sirtuin Modulators
WO2006094209A2 (en) N-benzimidazolylalkyl-substituted amide sirtuin modulators
US20100168084A1 (en) Therapeutic compounds and related methods of use
US20070149466A1 (en) Methods and related compositions for treating or preventing obesity, insulin resistance disorders, and mitochondrial-associated disorders
WO2006094210A2 (en) Tetrahydroquinoxalinone sirtuin modulators
WO2008073451A2 (en) Benzoimidazole derivatives as sirtuin (sir) modulating compounds
WO2006094246A2 (en) N-arylmethyl benzamide sirtuin modulators
EP1957109A2 (en) Modulators of cdc2-like kinases (clks) and methods of use thereof
CA2610854A1 (en) Nicotinamide riboside and analogues thereof
WO2006078941A2 (en) Novel sirtuin activating compounds and methods of use thereof
WO2006094233A1 (en) N,n'-dicyclic isothiourea sirtuin modulators
WO2009085226A2 (en) Inhibitors of cdc2-like kinases (clks) and methods of use thereof
WO2006094248A1 (en) Aryl-substituted cyclic sirtuin modulators
WO2009158646A1 (en) Therapeutic compunds and related methods of use

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20100728

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA RS

RIN1 Information on inventor provided before grant (corrected)

Inventor name: LAMBERT, PHILIP

Inventor name: ELLIOTT, PETER

Inventor name: LUNSMANN, WALTER, JOSEPH

RIN1 Information on inventor provided before grant (corrected)

Inventor name: LAMBERT, PHILIP

Inventor name: ELLIOTT, PETER

Inventor name: LUNSMANN, WALTER, JOSEPH

17Q First examination report despatched

Effective date: 20130328

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20130801